Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Front Bioeng Biotechnol ; 11: 1193430, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37324446

RESUMO

There is an urgent need to develop new therapies for colorectal cancer that has metastasized to the liver and, more fundamentally, to develop improved preclinical platforms of colorectal cancer liver metastases (CRCLM) to screen therapies for efficacy. To this end, we developed a multi-well perfusable bioreactor capable of monitoring CRCLM patient-derived organoid response to a chemotherapeutic gradient. CRCLM patient-derived organoids were cultured in the multi-well bioreactor for 7 days and the subsequently established gradient in 5-fluorouracil (5-FU) concentration resulted in a lower IC50 in the region near the perfusion channel versus the region far from the channel. We compared behaviour of organoids in this platform to two commonly used PDO culture models: organoids in media and organoids in a static (no perfusion) hydrogel. The bioreactor IC50 values were significantly higher than IC50 values for organoids cultured in media whereas only the IC50 for organoids far from the channel were significantly different than organoids cultured in the static hydrogel condition. Using finite element simulations, we showed that the total dose delivered, calculated using area under the curve (AUC) was similar between platforms, however normalized viability was lower for the organoid in media condition than in the static gel and bioreactor. Our results highlight the utility of our multi-well bioreactor for studying organoid response to chemical gradients and demonstrate that comparing drug response across these different platforms is nontrivial.

2.
PLoS One ; 18(4): e0271354, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37022995

RESUMO

Tumors rich in stroma are associated with advanced stage and poor prognosis in colorectal adenocarcinoma (CRC). Abundance of stromal cells also has implications for genomic analysis of patient tumors as it may prevent detection of somatic mutations. As part of our efforts to interrogate stroma-cancer cell interactions and to identify actionable therapeutic targets in metastatic CRC, we aimed to determine the proportion of stroma embedded in hepatic CRC metastases by performing computational tumor purity analysis based on whole exome sequencing data (WES). Unlike previous studies focusing on histopathologically prescreened samples, we used an unbiased in-house collection of tumor specimens. WES from CRC liver metastasis samples were utilized to evaluate stromal content and to assess the performance of three in silico tumor purity tools, ABSOLUTE, Sequenza and PureCN. Matching tumor derived organoids were analyzed as a high purity control as they are enriched in cancer cells. Computational purity estimates were compared to those from a histopathological assessment conducted by a board-certified pathologist. According to all computational methods, metastatic specimens had a median tumor purity of 30% whereas the organoids were enriched for cancer cells with a median purity estimate of 94%. In line with this, variant allele frequencies (VAFs) of oncogenes and tumor suppressor genes were undetectable or low in most patient tumors, but higher in matching organoid cultures. Positive correlation was observed between VAFs and in silico tumor purity estimates. Sequenza and PureCN produced concordant results whereas ABSOLUTE yielded lower purity estimates for all samples. Our data shows that unbiased sample selection combined with molecular, computational, and histopathological tumor purity assessment is critical to determine the level of stroma embedded in metastatic colorectal adenocarcinoma.


Assuntos
Adenocarcinoma , Neoplasias Colorretais , Neoplasias Hepáticas , Humanos , Sequenciamento do Exoma , Mutação , Exoma/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Adenocarcinoma/genética , Neoplasias Hepáticas/genética
3.
Microbiol Spectr ; 10(5): e0169422, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36154279

RESUMO

Enterovirus D68 (EV-D68) is an RNA virus that can cause outbreaks of acute flaccid paralysis (AFP), a polio-like disease. Before 2010, EV-D68 was a rare pathogen associated with mild respiratory symptoms, but the recent EV-D68 related increase in severe respiratory illness and outbreaks of AFP is not yet understood. An explanation for the rise in severe disease is that it may be due to changes in the viral genome resulting in neurotropism. In this regard, in addition to sialic acid, binding to heparan sulfate proteoglycans (HSPGs) has been identified as a feature for viral entry of some EV-D68 strains in cell lines. Studies in human primary organotypic cultures that recapitulate human physiology will address the relevance of these HSPG-binding mutations for EV-D68 infection in vivo. Therefore, in this work, we studied the replication and neurotropism of previously determined sialic acid-dependent and HSPG-dependent strains using primary human airway epithelial (HAE) cultures and induced human pluripotent stem cell (iPSC)-derived brain organoids. All three strains (B2/2042, B2/947, and A1/1348) used in this study infected HAE cultures and human brain organoids (shown for the first time). Receptor-blocking experiments in both cultures confirm that B2/2042 infection is solely dependent on sialic acid, while B2/947 and A1/1348 (HSPG to a lesser extent) binds to sialic acid and HSPG for cell entry. Our data suggest that HSPG-binding can be used by EV-D68 for entry in human physiological models but offers no advantage for EV-D68 infection of brain cells. IMPORTANCE Recent outbreaks of enterovirus D68, a nonpolio enterovirus, is associated with a serious neurological condition in young children, acute flaccid myelitis (AFM). As there is no antiviral treatment or vaccine available for EV-D68 it is important to better understand how EV-D68 causes AFM and why only recent outbreaks are associated with AFM. We investigated if a change in receptor usage of EV-D68 increases the virulence of EV-D68 in the airway or the central nervous system and thus could explain the increase in AFM cases. We studied this using physiologically relevant human airway epithelium and cerebral organoid cultures that are physiologically relevant human models. Our data suggest that heparan sulfate proteoglycans can be used by EV-D68 as an additional entry receptor in human physiological models but offers no advantage for EV-D68 infection of brain cells, and our data show the potential of these 46 innovative models for virology.


Assuntos
Enterovirus Humano D , Infecções por Enterovirus , Criança , Pré-Escolar , Humanos , Encéfalo/metabolismo , Enterovirus Humano D/genética , Infecções por Enterovirus/epidemiologia , Proteoglicanas de Heparan Sulfato/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Organoides
4.
J Cell Mol Med ; 26(16): 4591-4601, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35841281

RESUMO

Tumour Necrosis Factor (TNF) potently induces a transient inflammatory response that must be downregulated once any invasive stimulus has resolved. Yet, how TNF-induced inflammation is shut down in normal cells is incompletely understood. The present study shows that STAT3 was activated in mouse embryo fibroblasts (MEFs) by treatment with TNF or an agonist antibody to TNFR1. STAT3 activation was inhibited by pharmacological inhibition of the Jak2 tyrosine kinase that associates with TNFR1. To identify STAT3 target genes, global transcriptome analysis by RNA sequencing was performed in wild-type MEFs and MEFs from STAT3 knockout (STAT3KO ) mice that were stimulated with TNF, and the results were validated at the protein level by using multiplex cytokine assays and immunoblotting. After TNF stimulation, STAT3KO MEFs showed greater gene and protein induction of the inflammatory chemokines Ccl2, Cxcl1 and Cxcl10 than WT MEFs. These observations show that, by activating STAT3, TNF selectively modulates expression of a cohort of chemokines that promote inflammation. The greater induction by TNF of chemokines in STAT3KO than WT MEFs suggested that TNF induced an inhibitory protein in WT MEFs. Consistent with this possibility, STAT3 activation by TNFR1 increased the expression of Tnfaip3/A20, a ubiquitin modifying enzyme that inhibits inflammation, in WT MEFs but not in STAT3KO MEFs. Moreover, enforced expression of Tnfaip3/A20 in STAT3KO MEFs suppressed proinflammatory chemokine expression induced by TNF. Our observations identify Tnfaip3/A20 as a new downstream target for STAT3 which limits the induction of Ccl2, Cxcl1 and Cxcl10 and inflammation induced by TNF.


Assuntos
Receptores Tipo I de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa , Animais , Expressão Gênica , Inflamação , Janus Quinase 2/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Transcrição STAT3/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
5.
Viruses ; 11(11)2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31739613

RESUMO

ParechovirusA is a species in the Parechovirus genus within the Picornaviridae family that can cause severe disease in children. Relatively little is known on ParechovirusA epidemiology and pathogenesis. This review aims to explore the ParechovirusA literature and highlight the differences between ParechovirusA genotypes from a pathogenesis standpoint. In particular, the curious case of Parechovirus-A3 and the genotype-specific disease association will be discussed. Finally, a brief outlook on ParechovirusA research is provided.


Assuntos
Suscetibilidade a Doenças , Genótipo , Interações Hospedeiro-Patógeno , Parechovirus/classificação , Parechovirus/genética , Infecções por Picornaviridae/virologia , Gerenciamento Clínico , Evolução Molecular , Genoma Viral , Genômica/métodos , Humanos , Estágios do Ciclo de Vida , Parechovirus/crescimento & desenvolvimento , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/terapia
6.
Euro Surveill ; 24(35)2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31481149

RESUMO

BackgroundEnterovirus D68 (EV-D68) has caused major outbreaks of severe respiratory illness worldwide since 2010.AimOur aim was to evaluate EV-D68 circulation in the Netherlands by conducting a serosurvey of EV-D68 neutralising antibodies (nAb) among the Dutch general population.MethodsWe screened 280 sera from children and adults in the Netherlands and used two independent sets of samples collected in the years 2006 and 2007 and in the years 2015 and 2016, time points before and after the first EV-D68 upsurge in 2010. Neutralisation capacity of the sera was tested against the prototype Fermon EV-D68 strain isolated in 1962 and against a recent EV-D68 strain (genotype B3) isolated in France in 2016.ResultsRegardless of the time of serum collection, we found remarkably high overall seropositivity (94.3-98.3%) for nAb against both EV-D68 strains. Geometric mean titres increased in an age-dependent manner.ConclusionsOur data suggest that EV-D68 has been circulating in the Netherlands for decades and that the enterovirus surveillance does not accurately capture the prevalence of this clinically relevant pathogen.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Enterovirus Humano D/isolamento & purificação , Infecções por Enterovirus/epidemiologia , Infecções por Enterovirus/imunologia , Adolescente , Adulto , Distribuição por Idade , Idoso , Anticorpos Neutralizantes/análise , Criança , Pré-Escolar , Surtos de Doenças , Enterovirus Humano D/genética , Enterovirus Humano D/imunologia , Infecções por Enterovirus/sangue , Infecções por Enterovirus/diagnóstico , Humanos , Lactente , Pessoa de Meia-Idade , Países Baixos/epidemiologia , Testes de Neutralização , Prevalência , Estudos Soroepidemiológicos
7.
Arch Virol ; 164(3): 799-806, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30666460

RESUMO

Parechoviruses (PeVs) are highly prevalent viruses worldwide. Over the last decades, several studies have been published on PeV epidemiology in Europe, Asia and North America, while information on other continents is lacking. The aim of this study was to describe PeV circulation in a cohort of children in Malawi, Africa. A total of 749 stool samples obtained from Malawian children aged 6 to 60 months were tested for the presence of PeV by real-time PCR. We performed typing by phylogenetic and Basic Local Alignment Search Tool (BLAST) analysis. PeV was found in 57% of stool samples. Age was significantly associated with PeV positivity (p = 0.01). Typing by phylogenetic analysis resulted in 15 different types, while BLAST typing resulted in 14 different types and several indeterminate strains. In total, six strains showed inconsistencies in typing between the two methods. One strain, P02-4058, remained untypable by all methods, but appeared to belong to the recently reclassified PeV-A19 genotype. PeV-A1, -A2 and -A3 were the most prevalent types (26.8%, 13.8% and 9.8%, respectively). Both the prevalence and genetic diversity found in our study were remarkably high. Our data provide an important contribution to the scarce data available on PeV epidemiology in Africa.


Assuntos
Variação Genética , Parechovirus/isolamento & purificação , Infecções por Picornaviridae/virologia , Criança , Pré-Escolar , Estudos de Coortes , Fezes/virologia , Feminino , Genótipo , Humanos , Lactente , Malaui/epidemiologia , Masculino , Parechovirus/classificação , Parechovirus/genética , Filogenia , Infecções por Picornaviridae/epidemiologia
8.
Emerg Infect Dis ; 25(1): 148-152, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30561318

RESUMO

Recent parechovirus A3 (PeV-A3) outbreaks in Australia suggest lower population immunity compared with regions that have endemic PeV-A3 circulation. A serosurvey among populations in the Netherlands, the United States, and Australia before and after the 2013 Australia outbreak showed high PeV-A3 neutralizing antibody prevalence across all regions and time periods, indicating widespread circulation.


Assuntos
Anticorpos Antivirais/sangue , Surtos de Doenças , Parechovirus/imunologia , Infecções por Picornaviridae/epidemiologia , Adolescente , Adulto , Idoso , Anticorpos Neutralizantes/sangue , Austrália/epidemiologia , Criança , Pré-Escolar , Estudos Transversais , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Países Baixos/epidemiologia , Infecções por Picornaviridae/virologia , Estudos Soroepidemiológicos , Estados Unidos/epidemiologia , Adulto Jovem
9.
Pediatr Infect Dis J ; 37(12): 1304-1308, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30382954

RESUMO

BACKGROUND: Human parechoviruses (HPeVs) are common pathogens in young children, and in the Netherlands, HPeV1, HPeV3 and HPeV4 are the most frequently detected genotypes. HPeV3 in particular has been associated with severe disease in young infants below 3 months of age while the other genotypes more often infect older children and elicit mild symptoms. We investigated if maternal neutralizing antibodies (nAbs) against HPeV1, HPeV3 and HPeV4 protect young Dutch infants from severe disease related to HPeV infection. METHODS: We conducted a prospective case-control study of Dutch mother-infant pairs. Thirty-eight HPeV-infected infants and their mothers were included as cases, and 65 HPeV-negative children and their mothers as controls. RESULTS: In control infants, we observed nAb seropositivity rates of 41.4%, 33.3% and 27.6%, with median nAb titers of 1:16, 1:12 and 1:8, against HPeV1, HPeV3 and HPeV4, respectively. In control mothers, nAb seropositivity rates were 84.6%, 55.4% and 60.0% with median nAb titers of 1:128, 1:32 and 1:45 against HPeV1, HPeV3 and HPeV4, respectively. The HPeV3 nAb seroprevalence was significantly lower in HPeV3-infected infants and their mothers (0.0% with P < 0.05 and 10.0% with P < 0.001, respectively). In contrast, no differences in nAb seroprevalence against HPeV1 or HPeV4 could be detected between case and control infants or mothers. CONCLUSIONS: Our results suggest that young Dutch infants are protected against severe disease related to HPeV1 and HPeV4 by maternal nAbs, but less so against HPeV3 explaining the distinct age distributions and disease severity profiles of children infected with these HPeV genotypes.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/imunologia , Parechovirus/imunologia , Infecções por Picornaviridae/imunologia , Anticorpos Neutralizantes/imunologia , Estudos de Casos e Controles , Técnicas de Cultura de Células , Feminino , Genótipo , Humanos , Lactente , Masculino , Mães , Países Baixos , Parechovirus/genética , Estudos Prospectivos , Estudos Soroepidemiológicos
10.
Artigo em Inglês | MEDLINE | ID: mdl-30211126

RESUMO

Human parechoviruses (HPeVs), a poorly studied genus within the Picornaviridae family, are classified into 19 genotypes of which HPeV1 and HPeV3 are the most often detected. HPeV1 VP1 C terminus contains an arginine-glycine-aspartic acid (RGD) motif and has been shown to depend on the host cell surface αV integrins (αV ITGs) and heparan sulfate (HS) for entry. HPeV3 lacks this motif and the receptors remain unknown. HPeVs can be detected in patient nasopharyngeal and stool samples, and infection is presumed to occur after respiratory or gastro-intestinal transmission. HPeV pathogenesis is poorly understood as there are no animal models and previous studies have been conducted in immortalized monolayer cell cultures which do not adequately represent the characteristics of human tissues. To bridge this gap, we determined the polarity of infection, replication kinetics, and cell tropism of HPeV1 and HPeV3 in the well-differentiated human airway epithelial (HAE) model. We found the HAE cultures to be permissive for HPeVs. Both HPeV genotypes infected the HAE preferentially from the basolateral surface while the progeny virus was shed toward the apical side. Confocal microscopy revealed the target cell type to be the p63+ basal cells for both viruses, αV ITG and HS blocking had no effect on the replication of either virus, and transcriptional profiling suggested that HPeV3 infection induced stronger immune activation than HPeV1. Genotype-specific host responses may contribute to the differences in pathogenesis and clinical outcomes associated with HPeV1 and HPeV3.


Assuntos
Células Epiteliais/virologia , Epitélio/virologia , Parechovirus/fisiologia , Infecções por Picornaviridae/virologia , Internalização do Vírus , Polaridade Celular , Células Cultivadas , Humanos , Microscopia Confocal , Modelos Teóricos , Tropismo Viral , Liberação de Vírus , Replicação Viral
11.
Sci Rep ; 7(1): 12075, 2017 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-28935894

RESUMO

Human parechovirus 3 (HPeV3), a member of the Picornavirus family, is frequently detected worldwide. However, the observed seropositivity rates for HPeV3 neutralizing antibodies (nAbs) vary from high in Japan to low in the Netherlands and Finland. To study if this can be explained by technical differences or antigenic diversity among HPeV3 strains included in the serological studies, we determined the neutralizing activity of Japanese and Dutch intravenous immunoglobulin batches (IVIG), a rabbit HPeV3 hyperimmune polyclonal serum, and a human HPeV3-specific monoclonal antibody (mAb) AT12-015, against the HPeV3 A308/99 prototype strain and clinical isolates from Japan, the Netherlands and Australia, collected between 1989 and 2015. The rabbit antiserum neutralized all HPeV3 isolates whereas the neutralization capacity of the IVIG batches varied, and the mAb exclusively neutralized the A308/99 strain. Mapping of the amino acid variation among a subset of the HPeV3 strains on an HPeV3 capsid structure revealed that the majority of the surface-exposed amino acid variation was located in the VP1. Furthermore, amino acid mutations in a mAb AT12-015-resistant HPeV3 A308/99 variant indicated the location for potential antigenic determinants. Virus aggregation and the observed antigenic diversity in HPeV3 can explain the varying levels of nAb seropositivity reported in previous studies.


Assuntos
Anticorpos Neutralizantes/imunologia , Variação Antigênica/imunologia , Proteínas do Capsídeo/imunologia , Parechovirus/imunologia , Infecções por Picornaviridae/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/genética , Variação Antigênica/genética , Proteínas do Capsídeo/genética , Humanos , Soros Imunes/imunologia , Japão , Mutação , Países Baixos , Testes de Neutralização , Parechovirus/classificação , Parechovirus/fisiologia , Infecções por Picornaviridae/virologia , Coelhos , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
12.
Virol J ; 13(1): 171, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27756316

RESUMO

BACKGROUND: Coxsackievirus A9 (CV-A9) is a pathogenic enterovirus type within the family Picornaviridae. CV-A9 infects A549 human epithelial lung carcinoma cells by attaching to the αVß6 integrin receptor through a highly conserved Arg-Gly-Asp (RGD) motif, which is located at the exposed carboxy-terminus of the capsid protein VP1 detected in all studied clinical isolates. However, genetically-modified CV-A9 that lacks the RGD motif (CV-A9-RGDdel) has been shown to be infectious in some cell lines but not in A549, suggesting that RGD-mediated integrin binding is not always essential for efficient entry of CV-A9. METHODS: Two cell lines, A549 and SW480, were used in the study. SW480 was the study object for the integrin-independent entry and A549 was used as the control for integrin-dependent entry. Receptor levels were quantitated by cell sorting and quantitative PCR. Antibody blocking assay and siRNA silencing of receptor-encoding genes were used to block virus infection. Peptide phage display library was used to identify peptide binders to CV-A9. Immunofluorescence and confocal microscopy were used to visualize the virus infection in the cells. RESULTS: We investigated the receptor use and early stages of CV-A9 internalization to SW480 human epithelial colon adenocarcinoma cells. Contrary to A549 infection, we showed that both CV-A9 and CV-A9-RGDdel internalized into SW480 cells and that function-blocking anti-αV integrin antibodies had no effect on the binding and entry of CV-A9. Whereas siRNA silencing of ß6 integrin subunit had no influence on virus infection in SW480, silencing of ß2-microglobulin (ß2M) inhibited the virus infection in both cell lines. By using a peptide phage display screening, the virus-binding peptide identical to the N-terminal sequence of HSPA5 protein was identified and shown to block the virus infection in both A549 and SW480 cell lines. HSPA5 was also found to co-localize with CV-A9 at the SW480 cell periphery during the early stages of infection by confocal microscopy. CONCLUSIONS: The data suggest that while αVß6 integrin is essential for CV-A9 in A549 cell line, it is not required in SW480 cell line in which ß2M and HSPA5 alone are sufficient for CV-A9 infection. This suggests that the choice of CV-A9 receptor(s) is dependent on the tissue/cellular environment.


Assuntos
Antígenos de Neoplasias/metabolismo , Enterovirus Humano B/fisiologia , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Integrinas/metabolismo , Receptores Virais/metabolismo , Internalização do Vírus , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Humanos
13.
PLoS One ; 11(1): e0147168, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26785353

RESUMO

Heparan sulfate/heparin class of proteoglycans (HSPG) have been shown to function in cellular attachment and infection of numerous viruses including picornaviruses. Coxsackievirus A9 (CV-A9) and human parechovirus 1 (HPeV-1) are integrin-binding members in the family Picornaviridae. CV-A9 Griggs and HPeV-1 Harris (prototype) strains have been reported not to bind to heparin, but it was recently shown that some CV-A9 isolates interact with heparin in vitro via VP1 protein with a specific T132R/K mutation. We found that the infectivity of both CV-A9 Griggs and HPeV-1 Harris was reduced by sodium chlorate and heparinase suggestive of HSPG interactions. We analyzed the T132 site in fifty-four (54) CV-A9 clinical isolates and found that only one of them possessed T132/R mutation while the other nine (9) had T132K. We then treated CV-A9 Griggs and HPeV-1 Harris and eight CV-A9 and six HPeV-1 clinical isolates with heparin and protamine. Although infectivity of Griggs strain was slightly reduced (by 25%), heparin treatment did not affect the infectivity of the CV-A9 isolates that do not possess the T132R/K mutation, which is in line with the previous findings. Some of the HPeV-1 isolates were also affected by heparin treatment, which suggested that there may be a specific heparin binding site in HPeV-1. In contrast, protamine (a specific inhibitor of heparin) completely inhibited the infection of both prototypes and clinical CV-A9 and HPeV-1 isolates. We conclude that T132R/K mutation has a role in heparin binding of CV-A9, but we also show data, which suggest that there are other HSPG binding sites in CV-A9. In all, we suggest that HSPGs play a general role in both CV-A9 and HPeV-1 infections.


Assuntos
Infecções por Coxsackievirus/virologia , Enterovirus Humano B/isolamento & purificação , Heparitina Sulfato/metabolismo , Integrinas/metabolismo , Parechovirus/isolamento & purificação , Infecções por Picornaviridae/virologia , Sequência de Aminoácidos , Sítios de Ligação , Infecções por Coxsackievirus/metabolismo , Humanos , Dados de Sequência Molecular , N-Acetilglucosaminiltransferases/antagonistas & inibidores , N-Acetilglucosaminiltransferases/genética , Infecções por Picornaviridae/metabolismo , Homologia de Sequência de Aminoácidos , Proteínas Virais/metabolismo
14.
Growth Factors ; 32(6): 214-22, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25494541

RESUMO

Recombinant antibody phage library technology provides multiple advantages, including that human antibodies can be generated against proteins that are highly conserved between species. We used this technology to isolate and characterize an anti-EphA2 single-chain antibody. We show that the antibody binds the antigen with 1:1 stoichiometry and has high specificity for EphA2. The crystal structure of the complex reveals that the antibody targets the same receptor surface cavity as the ephrin ligand. Specifically, a lengthy CDR-H3 loop protrudes deep into the ligand-binding cavity, with several hydrophobic residues at its tip forming an anchor-like structure buried within the hydrophobic Eph pocket, in a way similar to the ephrin receptor-binding loop in the Eph/ephrin structures. Consequently, the antibody blocks ephrin binding to EphA2. Furthermore, it induces apoptosis and reduces cell proliferation in lymphoma cells lines. Since Ephs are important mediators of tumorigenesis, such antibodies could have applications both in research and therapy.


Assuntos
Receptor EphA2/imunologia , Anticorpos de Cadeia Única/imunologia , Sequência de Aminoácidos , Animais , Afinidade de Anticorpos , Sítios de Ligação de Anticorpos , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Humanos , Dados de Sequência Molecular , Anticorpos de Cadeia Única/química
15.
Protein Expr Purif ; 92(1): 112-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24036371

RESUMO

Eph receptors are the largest family of Receptor Tyrosine Kinases containing a single membrane-spanning segment. They are involved in a various developmental and cell-cell communication events. Although there is extensive structural information available on both the extra- and intracellular regions of Eph's in isolation, no structures are available for the entire receptor. To facilitate structural studies on functionally relevant Eph/ephrin complexes, we have developed an expression system for producing the full-length human EphA2 receptor. We successfully expressed milligram amounts of the receptor using baculovirus-based vector and insect cells. We were also able to extract the protein from the cell membranes and purify it to near homogeneity in two simple steps. The purified receptor was shown to retain its biological activity in terms of both binding to its functional ligands and being able to auto-phosphorylate the key tyrosine residues of the cytoplasmic kinase domain.


Assuntos
Clonagem Molecular/métodos , Receptor EphA2/química , Receptor EphA2/genética , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Linhagem Celular , Vetores Genéticos/genética , Humanos , Insetos , Dados de Sequência Molecular , Fosforilação , Receptor EphA2/isolamento & purificação , Receptor EphA2/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética
16.
Genome Announc ; 1(2): e0014613, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23580710

RESUMO

Genomes of three strains (Parker, USSR, and 275/58) of coxsackievirus A7 (CV-A7) were amplified by the long reverse transcription (RT)-PCR method and sequenced. While the sequences of Parker and USSR were identical, the similarities of 275/58 to the CV-A7 reference sequence, accession no. AY421765, were 82.6% and 96.2% for nucleotides and amino acids, respectively.

17.
Adv Virol ; 2012: 547530, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23227048

RESUMO

Picornaviruses that infect humans form one of the largest virus groups with almost three hundred virus types. They include significant enteroviral pathogens such as rhino-, polio-, echo-, and coxsackieviruses and human parechoviruses that cause wide range of disease symptoms. Despite the economic importance of picornaviruses, there are no antivirals. More than ten cellular receptors are known to participate in picornavirus infection, but experimental evidence of their role in cellular infection has been shown for only about twenty picornavirus types. Three enterovirus types and one parechovirus have experimentally been shown to bind and use integrin receptors in cellular infection. These include coxsackievirus A9 (CV-A9), echovirus 9, and human parechovirus 1 that are among the most common and epidemic human picornaviruses and bind to αV-integrins via RGD motif that resides on virus capsid. In contrast, echovirus 1 (E-1) has no RGD and uses integrin α2ß1 as cellular receptor. Endocytosis of CV-A9 has recently been shown to occur via a novel Arf6- and dynamin-dependent pathways, while, contrary to collagen binding, E-1 binds inactive ß1 integrin and enters via macropinocytosis. In this paper, we review what is known about receptors and endocytosis of integrin-binding human picornaviruses.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...