Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 95(9)2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33568504

RESUMO

Pestiviruses such as bovine viral diarrhea virus (BVDV) and classical swine fever virus (CSFV) belong to the family Flaviviridae and represent pathogens of outstanding veterinary relevance. Pestiviruses enter cells via receptor-mediated endocytosis. For entry in bovine cells, complement regulatory protein CD46bov serves as a cellular receptor for BVDV. In this study, the role of porcine CD46pig in cellular entry was investigated for the recently discovered atypical porcine pestivirus (APPV), CSFV, and Bungowannah virus (BuPV) in order to elucidate the observed differences in host cell tropism. A cell culture-adapted APPV variant, which shows enhanced viral replication in vitro, was generated and demonstrated a strict tropism of APPV for porcine cells. One of the porcine cell lines displayed areas of CD46pig-expressing cells and areas of nonexpressing cells, and one single cell line revealed not to express any CD46pig The CD46pig-deficient porcine lymphoma cell line, known to facilitate CSFV replication, was the only porcine cell line nonpermissive to APPV, indicating a significant difference in the entry mechanism of APPV and CSFV. Infection experiments with a set of genetically engineered CD46pig knockout cells confirmed that CD46pig is a major receptor of APPV as CD46bov is for BVDV. In contrast, it is apparently not an essential determinant in host cell entry of other porcine pestiviruses such as CSFV and BuPV. Existence of a CD46pig-independent entry mechanism illustrates that the pestiviral entry process is more diverse than previously recognized.IMPORTANCE Pestiviruses comprise animal pathogens such as classical swine fever virus (CSFV) and bovine viral diarrhea virus (BVDV) that cause notifiable diseases with great economic impact. Several additional pestivirus species affecting animal health were recently identified, including atypical porcine pestivirus (APPV). APPV is associated with health problems in piglets and is highly abundant in pig populations worldwide. Complement control protein CD46 serves as a receptor for diverse bacterial and viral pathogens, including particular adenoviruses, herpesviruses, measles virus (MeV), and BVDV. Porcine CD46 (CD46pig) was suggested to be a major receptor for CSFV. Here, we identified remarkable differences in relevance of CD46pig during entry of porcine pestiviruses. Resembling BVDV, efficient APPV infection in cell culture depends on CD46pig, while other porcine pestiviruses can efficiently enter and infect cells in the absence of CD46pig Thus, the study provides insights into the entry process of these pathogens and may help to understand differences in their biology.


Assuntos
Vírus da Febre Suína Clássica/fisiologia , Peste Suína Clássica/virologia , Proteína Cofatora de Membrana/fisiologia , Receptores Virais/fisiologia , Tropismo Viral , Internalização do Vírus , Animais , Linhagem Celular , Proteína Cofatora de Membrana/imunologia , Suínos
2.
Int J Surg ; 83: 184-188, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32987208

RESUMO

BACKGROUND: There are many causes of systemic complement activation, which may have detrimental effects on a pig xenograft. Transgenic expression of one or more human complement-regulatory proteins (hCRPs), e.g., hCD46, provides some protection to the xenograft, but it is not known whether it protects the xenograft from the effects of systemic complement activation. We used wild-type (WT) pig aortic endothelial cells (pAECs) to activate complement, and determined whether the expression of hCD46 on a1,3galactosyltransferase gene-knockout (GTKO) pAECs protected them from injury. METHODS: CFSE-labeled and non-labeled pAECs from a WT, a GTKO, or a GTKO/hCD46 pig were separately incubated with heat-inactivated pooled human serum in vitro. Antibody pre-bonded CFSE-labeled and non-labeled pAECs were mixed, and then incubated with rabbit complement. The complement-dependent cytotoxicity was measured by flow cytometry. RESULTS: There was significantly less lysis of GTKO/CD46 pAECs (6%) by 50% human serum compared to that of WT (91%, p<0.001) or GTKO (32%, p<0.01) pAECs. The lysis of GTKO pAECs was significantly increased when mixed with WT pAECs (p<0.05). In contrast, there was no significant change in cytotoxicity of GTKO/CD46 pAECs when mixed with WT pAECs. CONCLUSIONS: The expression of hCD46 protected pAECs from systemic complement activation.


Assuntos
Ativação do Complemento , Xenoenxertos/imunologia , Proteína Cofatora de Membrana/fisiologia , Animais , Animais Geneticamente Modificados , Aorta/imunologia , Citotoxicidade Imunológica , Células Endoteliais/imunologia , Humanos , Suínos
3.
Med Microbiol Immunol ; 209(3): 325-333, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31784892

RESUMO

Human Adenoviruses (HAdVs) are a family of clinically and therapeutically relevant viruses. A precise understanding of their host cell attachment and entry mechanisms can be applied in inhibitor design and the construction of targeted gene delivery vectors. In this article, structural data on adenovirus attachment and entry are reviewed. HAdVs engage two types of receptors: first, an attachment receptor that is bound by the fibre knob protein protruding from the icosahedral capsid, and next, an integrin entry receptor bound by the pentameric penton base at the capsid vertices. Adenoviruses use remarkably diverse attachment receptors, five of which have been studied structurally in the context of HAdV binding: Coxsackie and Adenovirus Receptor, CD46, the glycans GD1a and polysialic acid, and desmoglein-2. Together with the integrin entry receptors, they display both symmetrical and asymmetrical modes of binding to the virus as demonstrated by the structural analyses reviewed here. The diversity of HAdV receptors contributes to the broad tropism of these viruses, and structural studies are thus an important source of information on HAdV-host cell interactions. The imbalance in structural data between the more and less extensively studied receptors remains to be addressed by future research.


Assuntos
Adenovírus Humanos/fisiologia , Ligação Proteica , Receptores Virais/química , Receptores Virais/fisiologia , Ligação Viral , Internalização do Vírus , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/fisiologia , Desmogleína 2/fisiologia , Gangliosídeos/fisiologia , Interações entre Hospedeiro e Microrganismos , Humanos , Integrinas/fisiologia , Proteína Cofatora de Membrana/fisiologia , Ácidos Siálicos/fisiologia
4.
Front Immunol ; 10: 1074, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31164885

RESUMO

It has long been understood that the control and surveillance of tumors within the body involves an intricate dance between the adaptive and innate immune systems. At the center of the interplay between the adaptive and innate immune response sits the complement system-an evolutionarily ancient response that aids in the destruction of microorganisms and damaged cells, including cancer cells. Membrane-bound complement regulatory proteins (mCRPs), such as CD46, CD55, and CD59, are expressed throughout the body in order to prevent over-activation of the complement system. These mCRPs act as a double-edged sword however, as they can also over-regulate the complement system to the extent that it is no longer effective at eliminating cancerous cells. Recent studies are now indicating that mCRPs may function as a biomarker of a malignant transformation in numerous cancer types, and further, are being shown to interfere with anti-tumor treatments. This highlights the critical roles that therapeutic blockade of mCRPs can play in cancer treatment. Furthermore, with the complement system having the ability to both directly and indirectly control adaptive T-cell responses, the use of a combinatorial approach of complement-related therapy along with other T-cell activating therapies becomes a logical approach to treatment. This review will highlight the biomarker-related role that mCRP expression may have in the classification of tumor phenotype and predicted response to different anti-cancer treatments in the context of an emerging understanding that complement activation within the Tumor Microenvironment (TME) is actually harmful for tumor control. We will discuss what is known about complement activation and mCRPs relating to cancer and immunotherapy, and will examine the potential for combinatorial approaches of anti-mCRP therapy with other anti-tumor therapies, especially checkpoint inhibitors such as anti PD-1 and PD-L1 monoclonal antibodies (mAbs). Overall, mCRPs play an essential role in the immune response to tumors, and understanding their role in the immune response, particularly in modulating currently used cancer therapeutics may lead to better clinical outcomes in patients with diverse cancer types.


Assuntos
Antígenos CD55/fisiologia , Antígenos CD59/fisiologia , Imunoterapia/métodos , Proteína Cofatora de Membrana/fisiologia , Neoplasias/imunologia , Proteínas do Sistema Complemento/fisiologia , Humanos , Neoplasias/etiologia , Neoplasias/terapia , Receptores de Complemento 3b/fisiologia
5.
PLoS Pathog ; 15(2): e1007495, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30753248

RESUMO

The Gram-negative human pathogen N. gonorrhoeae (Ngo) quickly attaches to epithelial cells, and large numbers of the bacteria remain on the cell surface for prolonged periods. Ngo invades cells but few viable intracellular bacteria are recovered until later stages of infection, leading to the assumption that Ngo is a weak invader. On the cell surface, Ngo quickly recruits CD46-cyt1 to the epithelial cell cortex directly beneath the bacteria and causes its cleavage by metalloproteinases and Presenilin/γSecretease; how these interactions affect the Ngo lifecycle is unknown. Here, we show Ngo induces an autophagic response in the epithelial cell through CD46-cyt1/GOPC, and this response kills early invaders. Throughout infection, the pathogen slowly downregulates CD46-cyt1 and remodeling of lysosomes, another key autophagy component, and these activities ultimately promote intracellular survival. We present a model on the dynamics of Ngo infection and describe how this dual interference with the autophagic pathway allows late invaders to survive within the cell.


Assuntos
Gonorreia/metabolismo , Proteína Cofatora de Membrana/fisiologia , Neisseria gonorrhoeae/patogenicidade , Autofagia/fisiologia , Aderência Bacteriana , Linhagem Celular , Colo do Útero , Regulação para Baixo , Células Epiteliais , Feminino , Fímbrias Bacterianas , Gonorreia/fisiopatologia , Humanos , Lisossomos , Proteína Cofatora de Membrana/imunologia , Glicoproteínas de Membrana , Proteínas de Membrana/metabolismo , Neisseria gonorrhoeae/metabolismo , Cultura Primária de Células , Isoformas de Proteínas
6.
J Virol ; 93(3)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30429338

RESUMO

The impact of the Zika virus (ZIKV) epidemic highlights the need for vaccines that reduce or prevent infection and reliably prevent teratogenic complications. The live-attenuated measles virus (MV) vaccine strains are a promising vaccine platform, since they induce robust humoral and cellular immune responses against additional antigens and have an excellent safety record. To explore its potential to protect against ZIKV, we compared a recombinant Schwarz strain MV that encodes ZIKV prM and soluble E proteins (MV-Zika-sE) with a prototypic alum-adjuvanted whole inactivated ZIKV particle vaccine. Analysis of MV-Zika-sE-infected cells confirmed antigen expression, and the virus replicated with vaccine strain characteristics. Immunized IFNAR-/--CD46Ge mice developed E protein-specific and neutralizing antibodies, and ZIKV E-specific cellular immune responses were observed by gamma interferon (IFN-γ) enzyme-linked immunospot (ELISpot) and in vitro T cell proliferation assays. To analyze protective efficacy, vaccinated female mice were challenged with ZIKV after allogeneic mating. In MV-Zika-sE-vaccinated mice, weight gain was similar to that in uninfected mice, while no plasma viremia was detectable in the majority of the animals. In contrast, infected control animals gained less weight and experienced about 100-fold higher viremia over at least 3 days. Moreover, vaccination with MV-Zika-sE reduced the ZIKV load in different organs and the placentas and prevented infection of the fetus. Consequently, no fetal growth retardation, anemia, or death due to ZIKV infection was seen in MV-Zika-sE-vaccinated dams. In contrast, the inactivated ZIKV vaccine had little to no effect in our studies. Therefore, the MV-derived ZIKV vaccine is a promising candidate for further preclinical and clinical development.IMPORTANCE Zika virus (ZIKV) is a mosquito-borne flavivirus that causes a variety of neurological complications, including congenital birth defects. Despite the urgent need, no ZIKV vaccine has yet been licensed. Recombinant vaccine strain-derived measles viruses (MV) constitute a promising vector platform to induce immunity against foreign pathogens by expressing antigens from additional transcription units while at the same time possessing a remarkable safety profile. This concept has already been validated against different pathogens, including at least 3 other flaviviruses, and our data show that vaccination with MV expressing soluble ZIKV E protein significantly diminishes infection and prevents fetal loss or damage in an allogeneic mouse pregnancy model. It can thus be regarded as a promising emergency vaccine candidate with the potential for inclusion in routine vaccination settings in areas of endemicity to prevent teratogenic effects of circulating ZIKV during pregnancy, comparable to standard rubella virus vaccination.


Assuntos
Modelos Animais de Doenças , Vacina contra Sarampo/administração & dosagem , Vírus do Sarampo/imunologia , Proteínas do Envelope Viral/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/imunologia , Animais , Anticorpos Antivirais/sangue , Feminino , Genoma Viral , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Vacina contra Sarampo/imunologia , Proteína Cofatora de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Gravidez , Receptor de Interferon alfa e beta/fisiologia , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Zika virus/genética , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
7.
Hum Gene Ther Clin Dev ; 27(4): 145-151, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27604429

RESUMO

The modified Edmonston vaccine strain of measles virus (MV) has shown potent oncolytic efficacy against various tumor types and is being investigated in clinical trials. Our laboratory showed that MV effectively kills medulloblastoma tumor cells in both localized disease and when tumor cells are disseminated through cerebrospinal fluid (CSF). Although the safety of repeated intracerebral injection of modified MV in rhesus macaques has been established, the safety of administering MV into CSF has not been adequately investigated. In this study, we assessed the safety of MV-NIS (MV modified to express the human sodium iodide symporter protein) injected into the CSF of measles-immunized and measles virus-susceptible transgenic (CD46, IFNαRko) mice. Treated animals were administered a single intraventricular injection of 1 × 105 or 1 × 106 TCID50 (50% tissue culture infective dose) of MV-NIS. Detailed clinical observation was performed over a 90-day period. Clinically, we did not observe any measles-related toxic effects or behavioral abnormality in animals of any treated cohort. The complete blood count and blood chemistry analysis results were found to be within normal range for all the cohorts. Histologic examination of brains and spinal cords revealed inflammatory changes, mostly related to the needle track; these resolved by day 21 postinjection. To assess viral biodistribution, quantitative RT-PCR to detect the measles virus N-protein was performed on blood and brain samples. Viral RNA was not detectable in the blood as soon as 2 days after injection, and virus cleared from the brain by 45 days postadministration in all treatment cohorts. In conclusion, our data suggest that a single injection of modified MV into the CSF is safe and can be used in future therapeutic applications.


Assuntos
Vírus do Sarampo/patogenicidade , Sarampo/terapia , Proteína Cofatora de Membrana/fisiologia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Receptor de Interferon alfa e beta/fisiologia , Simportadores/fisiologia , Animais , Feminino , Humanos , Injeções Intraventriculares , Masculino , Sarampo/imunologia , Sarampo/virologia , Camundongos , Camundongos Transgênicos
8.
Eur J Cancer ; 50(12): 2152-61, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24915776

RESUMO

BACKGROUND: Membrane-bound complement restriction proteins (mCRPs) CD46, CD55 and CD59 enable tumour cells to evade complement dependent cytotoxicity and antibody-dependent killing mechanisms. But less is known about the role of these mCRPs in head and neck cancer. METHODS: In this study we determined the expression of the mCRPs on head and neck squamous cell carcinoma (HNSCC) cell lines, on tumour tissue and TDLNs (tumour-draining lymph nodes) as well as on lymphocytes from HNSCC patients. The influence of the HNSCC microenvironment on the mCRP regulation was analysed using Flow Cytometry, Western blotting and small interfering RNAs (siRNA) transfection studies. RESULTS: We examined the effects of the HNSCC tumour milieu on the expression levels of CD46, CD55 and CD59. We investigated the susceptibility of HNSCC cells to CDC (complement-dependent cytotoxicity) while silencing the mCRPs. Our results demonstrate a huge influence of the HNSCC tumour microenvironment on the regulation of mCRP expression and show a reciprocal regulation between the different mCRPs themselves. CONCLUSIONS: In summary, our data indicate that HNSCC has evolved different strategies to evade complement attacks and that the tumour microenvironment leads to the enhancement of complement resistance of the surrounding tissue.


Assuntos
Antígenos CD55/fisiologia , Antígenos CD59/fisiologia , Carcinoma de Células Escamosas/imunologia , Ativação do Complemento/fisiologia , Neoplasias de Cabeça e Pescoço/imunologia , Proteína Cofatora de Membrana/fisiologia , Neoplasias Bucais/imunologia , Microambiente Tumoral/fisiologia , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Feminino , Inativação Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma de Células Escamosas de Cabeça e Pescoço , Microambiente Tumoral/imunologia
10.
Pancreas ; 41(1): 84-94, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21934552

RESUMO

OBJECTIVES: There is a clear need for better therapeutics and diagnostics for pancreatic cancer. We aimed to discover plasma membrane-associated proteins overexpressed in pancreatic cancer using quantitative proteomics and apply RNA interference (RNAi) to uncover proteins associated with cancer cell survival. METHODS: Cell surface glycoproteins from 5 pancreatic cancer cell lines were isolated, and differential analyses were performed using mass spectrometry and the "normoid" cell line Hs766T as the comparator. For validation, immunohistochemistry was performed on tissues from 10 independent patients and 2 normal donors. Correlation of protein and mRNA expression level was determined, and functional activity characterized using RNAi. RESULTS: Integrin ß6, CD46, tissue factor, and a novel protein, chromosome 14 open reading frame 1, were identified as overexpressed on pancreatic cancer cell lines. Immunohistochemistry demonstrated the 4 targets were overexpressed in 20% to 70% of primary pancreatic tumor specimens. Small interfering RNA knockdown resulted in a reduction of cellular proliferation by inhibiting DNA synthesis, blocking S-phase progression or induction of apoptosis. CONCLUSIONS: By combining a mass spectrometry identification platform and an RNAi validation platform, we have identified a panel of cell surface glycoproteins that not only are overexpressed, but also play a functional role in pancreatic tumor cell survival.


Assuntos
Proliferação de Células , Glicoproteínas de Membrana/fisiologia , Proteômica/métodos , Interferência de RNA , Linhagem Celular Tumoral , Sobrevivência Celular , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Cadeias beta de Integrinas/genética , Cadeias beta de Integrinas/metabolismo , Cadeias beta de Integrinas/fisiologia , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/metabolismo , Proteína Cofatora de Membrana/fisiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tromboplastina/genética , Tromboplastina/metabolismo , Tromboplastina/fisiologia
12.
Infect Immun ; 78(9): 3832-47, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20584970

RESUMO

Neisseria meningitidis is a major cause of sepsis and meningitis but is also a common commensal, present in the nasopharynx of between 8 and 20% of healthy individuals. During carriage, the bacterium is found on the surface of the nasopharyngeal epithelium and in deeper tissues, while to develop disease the meningococcus must spread across the respiratory epithelium and enter the systemic circulation. Therefore, investigating the pathways by which N. meningitidis crosses the epithelial barrier is relevant for understanding carriage and disease but has been hindered by the lack of appropriate models. Here, we have established a physiologically relevant model of the upper respiratory epithelial cell barrier to investigate the mechanisms responsible for traversal of N. meningitidis. Calu-3 human respiratory epithelial cells were grown on permeable cell culture membranes to form polarized monolayers of cells joined by tight junctions. We show that the meningococcus crosses the epithelial cell barrier by a transcellular route; traversal of the layer did not disrupt its integrity, and bacteria were detected within the cells of the monolayer. We demonstrate that successful traversal of the epithelial cell barrier by N. meningitidis requires expression of its type 4 pili (Tfp) and capsule and is dependent on the host cell microtubule network. The Calu-3 model should be suitable for dissecting the pathogenesis of infections caused by other respiratory pathogens, as well as the meningococcus.


Assuntos
Neisseria meningitidis/fisiologia , Mucosa Respiratória/microbiologia , Aderência Bacteriana , Cápsulas Bacterianas/fisiologia , Células Cultivadas , Impedância Elétrica , Células Epiteliais/microbiologia , Humanos , Proteína Cofatora de Membrana/fisiologia , Proteínas de Membrana/análise , Microtúbulos/fisiologia , Fosfoproteínas/análise , Mucosa Respiratória/ultraestrutura , Proteína da Zônula de Oclusão-1
13.
J Biol Chem ; 285(27): 20882-90, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20435897

RESUMO

Measles virus (MV), an enveloped negative-strand RNA virus, remains a major cause of morbidity and mortality in developing countries. MV predominantly infects immune cells by using signaling lymphocyte activation molecule (SLAM; also called CD150) as a receptor, but it also infects polarized epithelial cells, forming tight junctions in a SLAM-independent manner. Although the ability of MV to infect polarized epithelial cells is thought to be important for its transmission, the epithelial cell receptor for MV has not been identified. A transcriptional repressor, Snail, induces epithelial-mesenchymal transition (EMT), in which epithelial cells lose epithelial cell phenotypes, such as adherens and tight junctions. In this study, EMT was induced by expressing Snail in a lung adenocarcinoma cell line, II-18, which is highly susceptible to wild-type MV. Snail-expressing II-18 cells lost adherens and tight junctions. Microarray analysis confirmed the induction of EMT in II-18 cells and suggested a novel function of Snail in protein degradation and distribution. Importantly, wild-type MV no longer entered EMT-induced II-18 cells, suggesting that the epithelial cell receptor is down-regulated by the induction of EMT. Other polarized cell lines, NCI-H358 and HT-29, also lost susceptibility to wild-type MV when EMT was induced. However, the complete formation of tight junctions rather reduced MV entry into HT-29 cells. Taken together, these data suggest that the unidentified epithelial cell receptor for MV is involved in the formation of epithelial intercellular junctions.


Assuntos
Células Epiteliais/citologia , Vírus do Sarampo/patogenicidade , Sarampo/prevenção & controle , Mesoderma/citologia , Animais , Linhagem Celular , Suscetibilidade a Doenças , Células Epiteliais/fisiologia , Células Epiteliais/virologia , Citometria de Fluxo , Predisposição Genética para Doença , Vetores Genéticos , Genoma Viral , Humanos , Ativação Linfocitária , Macaca mulatta/virologia , Sarampo/imunologia , Sarampo/transmissão , Sarampo/veterinária , Vírus do Sarampo/genética , Proteína Cofatora de Membrana/fisiologia , Mesoderma/fisiologia , Mesoderma/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos , Receptores Virais/fisiologia , Junções Íntimas/fisiologia , Eliminação de Partículas Virais
14.
J Virol ; 84(10): 5336-50, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20237079

RESUMO

Human adenovirus serotype 35 (HAdV-35; here referred to as Ad35) causes kidney and urinary tract infections and infects respiratory organs of immunocompromised individuals. Unlike other adenoviruses, Ad35 has a low seroprevalence, which makes Ad35-based vectors promising candidates for gene therapy. Ad35 utilizes CD46 and integrins as receptors for infection of epithelial and hematopoietic cells. Here we show that infectious entry of Ad35 into HeLa cells, human kidney HK-2 cells, and normal human lung fibroblasts strongly depended on CD46 and integrins but not heparan sulfate and variably required the large GTPase dynamin. Ad35 infections were independent of expression of the carboxy-terminal domain of AP180, which effectively blocks clathrin-mediated uptake. Ad35 infections were inhibited by small chemicals against serine/threonine kinase Pak1 (p21-activated kinase), protein kinase C (PKC), sodium-proton exchangers, actin, and acidic organelles. Remarkably, the F-actin inhibitor jasplakinolide, the Pak1 inhibitor IPA-3, or the sodium-proton exchange inhibitor 5-(N-ethyl-N-isopropyl) amiloride (EIPA) blocked endocytic uptake of Ad35. Dominant-negative proteins or small interfering RNAs against factors driving macropinocytosis, including the small GTPase Rac1, Pak1, or the Pak1 effector C-terminal binding protein 1 (CtBP1), potently inhibited Ad35 infection. Confocal laser scanning microscopy, electron microscopy, and live cell imaging showed that Ad35 colocalized with fluid-phase markers in large endocytic structures that were positive for CD46, alphanu integrins, and also CtBP1. Our results extend earlier observations with HAdV-3 (Ad3) and establish macropinocytosis as an infectious pathway for species B human adenoviruses in epithelial and hematopoietic cells.


Assuntos
Adenovírus Humanos/fisiologia , Células Epiteliais/virologia , Pinocitose , Internalização do Vírus , Linhagem Celular , Fibroblastos/virologia , Humanos , Integrinas/fisiologia , Proteína Cofatora de Membrana/fisiologia , Receptores Virais/fisiologia
15.
J Gen Virol ; 91(Pt 4): 971-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19923259

RESUMO

The lymphotropic and myelotropic nature of wild-type measles virus (wt-MV) is well recognized, with dendritic cells and lymphocytes expressing the MV receptor CD150 mediating systemic spread of the virus. Infection of respiratory epithelial cells has long been considered crucial for entry of MV into the body. However, the lack of detectable CD150 on these cells raises the issue of their importance in the pathogenesis of measles. This study utilized a combination of in vitro, ex vivo and in vivo model systems to characterize the susceptibility of epithelial cells to wt-MV of proven pathogenicity. Low numbers of MV-infected epithelial cells in close proximity to underlying infected lymphocytes or myeloid cells suggested infection via the basolateral side of the epithelium in the macaque model. In primary cultures of human bronchial epithelial cells, foci of MV-infected cells were only observed following infection via the basolateral cell surface. The extent of infection in primary cells was enhanced both in vitro and in ex vivo cornea rim tissue by disrupting the integrity of the cells prior to the application of virus. This demonstrated that, whilst epithelial cells may not be the primary target cells for wt-MV, areas of epithelium in which tight junctions are disrupted can become infected using high m.o.i. The low numbers of MV-infected epithelial cells observed in vivo in conjunction with the absence of infectious virus release from infected primary cell cultures suggest that epithelial cells have a peripheral role in MV transmission.


Assuntos
Células Gigantes/fisiologia , Vírus do Sarampo/fisiologia , Liberação de Vírus , Animais , Brônquios/virologia , Células Cultivadas , Chlorocebus aethiops , Células Epiteliais/virologia , Proteínas de Fluorescência Verde , Humanos , Macaca , Proteína Cofatora de Membrana/fisiologia , Células Vero , Internalização do Vírus
16.
Thromb Haemost ; 102(6): 1050-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19967134

RESUMO

As the major entry receptor, signalling lymphocytic activation molecule (SLAM, CD150) essentially determines the tropism of measles virus (MV) for immune cells. This receptor is of considerable importance for the induction of immunomodulation and -suppression, and for the systemic spread of MV to organs including secondary lymphoid tissues, the skin, the respiratory tract, and the brain predominantly via infected cells of the immune system. But how does the virus cross the epithelial barrier during initiation of the infection, the blood organ barriers formed by endothelial cells, and the epithelial barrier from within, when virus will be released from the host? Additional unknown receptor(s) on CD150-negative epithelial and endothelial cells have been postulated. However, it has also been postulated (and demonstrated in macaques) that the initial infection is independent from usage of this receptor, and that the first target cells appear to be CD150-positive cells in the epithelium. For later stages of the infection, for virus release from the host, it is claimed that this unknown receptor on epithelial cells is required for crossing the barrier from within. The endothelial cell barrier must be crossed from the apical (luminal) to the basolateral (abluminal) side to carry the infection to organs and the skin. However, infected leukocytes are impaired in several functions including transmigration through endothelial cells. The infection may spread via cell contact-mediated infection of endothelial cells and basolateral virus release, or via migration of infected leukocytes.


Assuntos
Vírus do Sarampo/patogenicidade , Sarampo/virologia , Animais , Antígenos CD/fisiologia , Modelos Animais de Doenças , Endotélio Vascular/imunologia , Endotélio Vascular/virologia , Epitélio/imunologia , Epitélio/virologia , Interações Hospedeiro-Patógeno , Humanos , Sarampo/etiologia , Sarampo/imunologia , Vírus do Sarampo/imunologia , Vírus do Sarampo/fisiologia , Proteína Cofatora de Membrana/fisiologia , Modelos Biológicos , Receptores de Superfície Celular/fisiologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Proteínas do Envelope Viral/fisiologia
17.
Oncology ; 77(6): 366-77, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20068366

RESUMO

Adenoviral vector-mediated transfer of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can be a powerful approach to lung cancer therapy. However, the efficiency of adenoviral vector gene transfer and the sensitivity to TRAIL-induced apoptosis in the context of adenoviral vector gene transfer have yet to be characterized in primary lung cancers. In this study, we investigated the expression of adenoviral receptor CD46 expression in primary lung cancer cells. In contrast to previous reports on enhanced CD46 expression in various types of cancer cells, we show a significantly higher CD46 expression in lung adenocarcinomas compared to lung squamous cell carcinomas. Using Ad5-GFP and Ad5F35-GFP vectors, we demonstrated an improved gene transfer efficiency in primary lung cancer cells by the Ad5F35 vector. The apoptosis induction effect mediated by Ad5-TRAIL and Ad5F35-TRAIL vector gene transfer was compared in cells from 10 lung adenocarcinomas. Of 5 lung cancers in which apoptosis was induced, 2 had an enhanced effect by Ad5F35-TRAIL vector gene transfer compared to Ad5-GFP. Thus, these results indicate a method to identify TRAIL-sensitive primary lung cancers, which will also facilitate the analysis of resistance mechanisms in lung cancers.


Assuntos
Adenocarcinoma/terapia , Adenoviridae/genética , Terapia Genética , Neoplasias Pulmonares/terapia , Proteína Cofatora de Membrana/fisiologia , Receptores Virais/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Linhagem Celular Tumoral , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Feminino , Vetores Genéticos , Humanos , Masculino , Pessoa de Meia-Idade
18.
J Immunol ; 181(4): 2544-55, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18684945

RESUMO

Tissue homing of activated T cells is typically mediated through their specific integrin and chemokine receptor repertoire. Activation of human primary CD4(+) T cells in the presence of CD46 cross-linking induces the development of a distinct immunomodulatory T cell population characterized by high IL-10/granzyme B production. How these regulatory T cells (Tregs) migrate/home to specific tissue sites is not understood. In this study, we determined the adhesion protein and chemokine receptor expression pattern on human CD3/CD46-activated peripheral blood CD4(+) T cells. CD3/CD46-activated, but not CD3/CD28-activated, T cells up-regulate the integrin alpha(4)beta(7). The interaction of alpha(4)beta(7) with its ligand mucosal addressin cell adhesion molecule 1 (MAdCAM-1) mediates homing or retention of T cells to the intestine. CD3/CD46-activated Tregs adhere to/roll on MAdCAM-1-expressing HeLa cells, similar to T cells isolated from the human lamina propria (LP). This interaction is inhibited by silencing MAdCAM-1 expression in HeLa cells or by the addition of blocking Abs to beta(7). CD46 activation of T cells also induced the expression of the surface-bound cytokine LIGHT and the chemokine receptor CCR9, both marker constitutively expressed by gut LP-resident T cells. In addition, we found that approximately 10% of the CD4(+) T lymphocytes isolated from the LP of patients undergoing bariatric surgery contain T cells that spontaneously secrete a cytokine pattern consistent with that from CD46-activated T cells. These data suggest that CD46-induced Tregs might play a role in intestinal immune homeostasis where they could dampen unwanted effector T cell responses through local IL-10/granzyme B production.


Assuntos
Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Ativação Linfocitária/imunologia , Proteína Cofatora de Membrana/fisiologia , Receptores de Quimiocinas/biossíntese , Receptores de Retorno de Linfócitos/biossíntese , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Moléculas de Adesão Celular , Células Cultivadas , Quimiotaxia de Leucócito/imunologia , Granzimas/biossíntese , Granzimas/fisiologia , Células HeLa , Humanos , Imunoglobulinas/biossíntese , Imunoglobulinas/fisiologia , Integrinas/biossíntese , Integrinas/fisiologia , Interleucina-10/biossíntese , Interleucina-10/metabolismo , Mucosa Intestinal/citologia , Mucoproteínas/biossíntese , Mucoproteínas/fisiologia , Receptores de Quimiocinas/genética , Receptores de Retorno de Linfócitos/fisiologia , Regulação para Cima/imunologia
19.
Ai Zheng ; 25(11): 1450-3, 2006 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-17094920

RESUMO

Immunologic mechanism of tumor escaping from complements' attack was unclear in the past. The efficacy of immunotherapy, especially humoral immunotherapy, to tumor is unsatisfactory. At present, with the progression in immunology, various tumors were found to highly express one or several kinds of complement regulatory proteins, such as CD46/MCP, CD55/DAF, and CD59/potectin. Complement system of the organism is inhibited because of high expression of complement regulatory proteins; therefore, the tumor can escape from the attack of complement system. Recently, the mechanism of complement regulatory proteins expressing in tumor has been studied in deep; some immunotherapies aim directly at complement regulatory proteins, including monoclonal antibody of complement regulatory proteins and cytokines, have been applied to animal experiments and clinical trails, and got some success. This review elucidated the progression on complement regulatory proteins in tumor immunotherapy.


Assuntos
Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Imunoterapia , Proteína Cofatora de Membrana/metabolismo , Neoplasias/terapia , Animais , Antígenos CD55/fisiologia , Antígenos CD59/fisiologia , Humanos , Interleucina-4/farmacologia , Proteína Cofatora de Membrana/fisiologia , Neoplasias/imunologia
20.
J Immunol ; 177(8): 4957-61, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17015676

RESUMO

T regulatory cell 1 (Tr1) are low proliferating peripherally induced suppressive T cells. Engaging CD3 and CD46 on human CD4+ T cells induces a Tr1-like phenotype. In this study, we report that human Tr1-like cells do not sustain proliferation over time. The weak proliferation of these cells results first from their inability to sustain expression of various cell cycle-associated proteins, to efficiently degrade the inhibitor of cell cycle progression p27/Kip1 and, as a consequence, in their accumulation in the G0-G1 phase. Also, the reduced proliferation of Tr1-like cells results from their increased sensitivity to death as they divide, through a mechanism that is neither Fas-mediated nor Bcl2/Bcl-xL related. Both properties, impaired cell cycle and death sensitivity, are explained by a specific defective activation of Akt that impairs the expression of Survivin. Thus, our results show that CD3/CD46-induced Tr1-like cells die through a process of abortive proliferation.


Assuntos
Proliferação de Células , Proteína Cofatora de Membrana/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais , Linfócitos T Reguladores/citologia , Complexo CD3/fisiologia , Ciclo Celular , Morte Celular , Inibidor de Quinase Dependente de Ciclina p27 , Humanos , Proteínas Inibidoras de Apoptose , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Survivina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA