Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 95(9)2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33568504

RESUMEN

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.


Asunto(s)
Virus de la Fiebre Porcina Clásica/fisiología , Peste Porcina Clásica/virología , Proteína Cofactora de Membrana/fisiología , Receptores Virales/fisiología , Tropismo Viral , Internalización del Virus , Animales , Línea Celular , Proteína Cofactora de Membrana/inmunología , Porcinos
2.
PLoS Pathog ; 15(2): e1007495, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30753248

RESUMEN

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.


Asunto(s)
Gonorrea/metabolismo , Proteína Cofactora de Membrana/fisiología , Neisseria gonorrhoeae/patogenicidad , Autofagia/fisiología , Adhesión Bacteriana , Línea Celular , Cuello del Útero , Regulación hacia Abajo , Células Epiteliales , Femenino , Fimbrias Bacterianas , Gonorrea/fisiopatología , Humanos , Lisosomas , Proteína Cofactora de Membrana/inmunología , Glicoproteínas de Membrana , Proteínas de la Membrana/metabolismo , Neisseria gonorrhoeae/metabolismo , Cultivo Primario de Células , Isoformas de Proteínas
3.
J Virol ; 93(3)2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30429338

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Vacuna Antisarampión/administración & dosificación , Virus del Sarampión/inmunología , Proteínas del Envoltorio Viral/inmunología , Infección por el Virus Zika/prevención & control , Virus Zika/inmunología , Animales , Anticuerpos Antivirales/sangre , Femenino , Genoma Viral , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Vacuna Antisarampión/inmunología , Proteína Cofactora de Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Embarazo , Receptor de Interferón alfa y beta/fisiología , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Virus Zika/genética , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
4.
Med Microbiol Immunol ; 209(3): 325-333, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31784892

RESUMEN

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.


Asunto(s)
Adenovirus Humanos/fisiología , Unión Proteica , Receptores Virales/química , Receptores Virales/fisiología , Acoplamiento Viral , Internalización del Virus , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/fisiología , Desmogleína 2/fisiología , Gangliósidos/fisiología , Interacciones Microbiota-Huesped , Humanos , Integrinas/fisiología , Proteína Cofactora de Membrana/fisiología , Ácidos Siálicos/fisiología
5.
J Virol ; 86(18): 9929-40, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22761385

RESUMEN

Enveloped viruses can incorporate host cell membrane proteins during the budding process. Here we demonstrate that mumps virus (MuV) and vesicular stomatitis virus (VSV) assemble to include CD46 and CD55, two host cell regulators which inhibit propagation of complement pathways through distinct mechanisms. Using viruses which incorporated CD46 alone, CD55 alone, or both CD46 and CD55, we have tested the relative contribution of these regulators in resistance to complement-mediated neutralization. Virion-associated CD46 and CD55 were biologically active, with VSV showing higher levels of activity of both cofactors, which promoted factor I-mediated cleavage of C3b into iC3b as well as decay-accelerating factor (DAF) activity against the C3 convertase, than MuV. Time courses of in vitro neutralization with normal human serum (NHS) showed that both regulators could delay neutralization, but viruses containing CD46 alone were neutralized faster and more completely than viruses containing CD55 alone. A dominant inhibitory role for CD55 was most evident for VSV, where virus containing CD55 alone was not substantially different in neutralization kinetics from virus harboring both regulators. Electron microscopy showed that VSV neutralization proceeded through virion aggregation followed by lysis, with virion-associated CD55 providing a delay in both aggregation and lysis more substantial than that conferred by CD46. Our results demonstrate the functional significance of incorporation of host cell factors during virion envelope assembly. They also define pathways of virus complement-mediated neutralization and suggest the design of more effective viral vectors.


Asunto(s)
Antígenos CD55/fisiología , Activación de Complemento/fisiología , Proteína Cofactora de Membrana/fisiología , Virus de la Parotiditis/inmunología , Vesiculovirus/inmunología , Animales , Antígenos CD55/genética , Células CHO , Activación de Complemento/genética , Cricetinae , Cricetulus , Interacciones Huésped-Patógeno/inmunología , Humanos , Proteína Cofactora de Membrana/genética , Microscopía Inmunoelectrónica , Virus de la Parotiditis/fisiología , Virus de la Parotiditis/ultraestructura , Pruebas de Neutralización , Vesiculovirus/fisiología , Vesiculovirus/ultraestructura , Ensamble de Virus
6.
Adv Exp Med Biol ; 735: 55-81, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23402019

RESUMEN

For the last two decades, there had been remarkable advancement in understanding the role of complement regulatory proteins in autoimmune disorders and importance of complement inhibitors as therapeutics. Systemic lupus erythematosus is a prototype of systemic autoimmune disorders. The disease, though rare, is potentially fatal and afflicts women at their reproductive age. It is a complex disease with multiorgan involvement, and each patient presents with a different set of symptoms. The diagnosis is often difficult and is based on the diagnostic criteria set by the American Rheumatology Association. Presence of antinuclear antibodies and more specifically antidouble-stranded DNA indicates SLE. Since the disease is multifactorial and its phenotypes are highly heterogeneous, there is a need to identify multiple noninvasive biomarkers for SLE. Lack of validated biomarkers for SLE disease activity or response to treatment is a barrier to the efficient management of the disease, drug discovery, as well as development of new therapeutics. Recent studies with gene knockout mice have suggested that membrane-bound complement regulatory proteins (CRPs) may critically determine the sensitivity of host tissues to complement injury in autoimmune and inflammatory disorders. Case-controlled and followup studies carried out in our laboratory suggest an intimate relation between the level of DAF, MCP, CR1, and CD59 transcripts and the disease activity in SLE. Based on comparative evaluation of our data on these four membrane-bound complement regulatory proteins, we envisaged CR1 and MCP transcripts as putative noninvasive disease activity markers and the respective proteins as therapeutic targets for SLE. Following is a brief appraisal on membrane-bound complement regulatory proteins DAF, MCP, CR1, and CD59 as biomarkers and therapeutic targets for SLE.


Asunto(s)
Proteínas del Sistema Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/fisiología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/fisiología , Animales , Biomarcadores , Antígenos CD55/fisiología , Antígenos CD59/efectos de los fármacos , Antígenos CD59/fisiología , Humanos , Proteína Cofactora de Membrana/antagonistas & inhibidores , Proteína Cofactora de Membrana/fisiología , Receptores de Complemento/antagonistas & inhibidores , Receptores de Complemento/fisiología
7.
J Immunol ; 184(2): 694-701, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20018629

RESUMEN

CD46 is a type I transmembrane protein with complement and T cell regulatory functions in human cells. CD46 has signaling and receptor properties in immune and nonimmune cells, many of which are dependent on the expression of cytoplasmic tail (cyt) isoforms cyt1 or cyt2. Little is known about how cyt1 and cyt2 mediate cellular responses. We show that CD46-cyt1 and CD46-cyt2 are substrates for presenilin/gamma-secretase (PS/gammaS), an endogenous protease complex that regulates many important signaling proteins through proteolytic processing. PS/gammaS processing of CD46 releases immunoprecipitable cyt1 and cyt2 tail peptides into the cell, is blocked by chemical inhibitors, and is prevented in dominant negative presenilin mutant cell lines. Two human pathogens, Neisseria gonorrhoeae and Neisseria meningitidis, stimulate PS/gammaS processing of CD46-cyt1 and CD46-cyt2. This stimulation requires type IV pili and PilT, the type IV pilus retraction motor, implying that mechanotransduction plays a role in this event. We present a model for PS/gammaS processing of CD46 that provides a mechanism by which signals are transduced via the cyt1 and cyt2 tails to regulate CD46-dependent cellular responses. Our findings have broad implications for understanding the full range of CD46 functions in infection and noninfection situations.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Gonorrea/metabolismo , Proteína Cofactora de Membrana/metabolismo , Infecciones Meningocócicas/metabolismo , Presenilinas/metabolismo , Fimbrias Bacterianas , Humanos , Mecanotransducción Celular , Proteína Cofactora de Membrana/fisiología , Neisseria gonorrhoeae , Neisseria meningitidis , Isoformas de Proteínas , Transducción de Señal
8.
J Biol Chem ; 285(27): 20882-90, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20435897

RESUMEN

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.


Asunto(s)
Células Epiteliales/citología , Virus del Sarampión/patogenicidad , Sarampión/prevención & control , Mesodermo/citología , Animales , Línea Celular , Susceptibilidad a Enfermedades , Células Epiteliales/fisiología , Células Epiteliales/virología , Citometría de Flujo , Predisposición Genética a la Enfermedad , Vectores Genéticos , Genoma Viral , Humanos , Activación de Linfocitos , Macaca mulatta/virología , Sarampión/inmunología , Sarampión/transmisión , Sarampión/veterinaria , Virus del Sarampión/genética , Proteína Cofactora de Membrana/fisiología , Mesodermo/fisiología , Mesodermo/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Plásmidos , Receptores Virales/fisiología , Uniones Estrechas/fisiología , Esparcimiento de Virus
9.
J Virol ; 84(10): 5336-50, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20237079

RESUMEN

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.


Asunto(s)
Adenovirus Humanos/fisiología , Células Epiteliales/virología , Pinocitosis , Internalización del Virus , Línea Celular , Fibroblastos/virología , Humanos , Integrinas/fisiología , Proteína Cofactora de Membrana/fisiología , Receptores Virales/fisiología
10.
J Virol ; 84(7): 3413-20, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20071568

RESUMEN

The signaling lymphocytic activation molecule (SLAM; CD150) is the immune cell receptor for measles virus (MV). To assess the importance of the SLAM-MV interactions for virus spread and pathogenesis, we generated a wild-type IC-B MV selectively unable to recognize human SLAM (SLAM-blind). This virus differs from the fully virulent wild-type IC-B strain by a single arginine-to-alanine substitution at amino acid 533 of the attachment protein hemagglutinin and infects cells through SLAM about 40 times less efficiently than the isogenic wild-type strain. Ex vivo, this virus infects primary lymphocytes at low levels regardless of SLAM expression. When a group of six rhesus monkeys (Macaca mulatta) was inoculated intranasally with the SLAM-blind virus, no clinical symptoms were documented. Only one monkey had low-level viremia early after infection, whereas all the hosts in the control group had high viremia levels. Despite minimal, if any, viremia, all six hosts generated neutralizing antibody titers close to those of the control monkeys while MV-directed cellular immunity reached levels at least as high as in wild-type-infected monkeys. These findings prove formally that efficient SLAM recognition is necessary for MV virulence and pathogenesis. They also suggest that the selectively SLAM-blind wild-type MV can be developed into a vaccine vector.


Asunto(s)
Inmunidad Adaptativa , Antígenos CD/fisiología , Virus del Sarampión/fisiología , Receptores de Superficie Celular/fisiología , Animales , Línea Celular , Humanos , Macaca mulatta , Vacuna Antisarampión/inmunología , Virus del Sarampión/inmunología , Virus del Sarampión/patogenicidad , Proteína Cofactora de Membrana/fisiología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Linfocitos T/virología , Proteínas Virales/fisiología , Internalización del Virus
11.
Infect Immun ; 78(9): 3832-47, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20584970

RESUMEN

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.


Asunto(s)
Neisseria meningitidis/fisiología , Mucosa Respiratoria/microbiología , Adhesión Bacteriana , Cápsulas Bacterianas/fisiología , Células Cultivadas , Impedancia Eléctrica , Células Epiteliales/microbiología , Humanos , Proteína Cofactora de Membrana/fisiología , Proteínas de la Membrana/análisis , Microtúbulos/fisiología , Fosfoproteínas/análisis , Mucosa Respiratoria/ultraestructura , Proteína de la Zonula Occludens-1
12.
J Gen Virol ; 91(Pt 4): 971-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19923259

RESUMEN

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.


Asunto(s)
Células Gigantes/fisiología , Virus del Sarampión/fisiología , Liberación del Virus , Animales , Bronquios/virología , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/virología , Proteínas Fluorescentes Verdes , Humanos , Macaca , Proteína Cofactora de Membrana/fisiología , Células Vero , Internalización del Virus
13.
J Immunol ; 181(4): 2544-55, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18684945

RESUMEN

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.


Asunto(s)
Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Activación de Linfocitos/inmunología , Proteína Cofactora de Membrana/fisiología , Receptores de Quimiocina/biosíntesis , Receptores Mensajeros de Linfocitos/biosíntesis , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Moléculas de Adhesión Celular , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Granzimas/biosíntesis , Granzimas/fisiología , Células HeLa , Humanos , Inmunoglobulinas/biosíntesis , Inmunoglobulinas/fisiología , Integrinas/biosíntesis , Integrinas/fisiología , Interleucina-10/biosíntesis , Interleucina-10/metabolismo , Mucosa Intestinal/citología , Mucoproteínas/biosíntesis , Mucoproteínas/fisiología , Receptores de Quimiocina/genética , Receptores Mensajeros de Linfocitos/fisiología , Regulación hacia Arriba/inmunología
14.
Uirusu ; 60(2): 221-35, 2010 Dec.
Artículo en Japonés | MEDLINE | ID: mdl-21488335

RESUMEN

human herpesvirus 6 (HHV-6) is the major causative agent of exanthem subitum which is one of popular diseases in infant, and establishes latent infections in adults of more than 90%. Recently, the encephalitis caused by reactivated- HHV-6 has been shown in patients after transplantation. In addition, the relationship HHV-6 and drug-induced hypersensitivity syndrome has also been reported. human herpesvirus 7 (HHV-7) was isolated from the stimulated-peripheral blood lymphocytes of a healthy individual, and also causes exanthema subitum. Both viruses are related viruses which belong to betaherpesvirus subfamily, and replicate and produce progeny viruses in T cells.


Asunto(s)
Exantema Súbito , Herpesvirus Humano 6 , Herpesvirus Humano 7 , Adulto , Exantema Súbito/diagnóstico , Exantema Súbito/terapia , Exantema Súbito/transmisión , Exantema Súbito/virología , Regulación Viral de la Expresión Génica , Genes Virales/genética , Genoma Viral/genética , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/inmunología , Herpesvirus Humano 6/patogenicidad , Herpesvirus Humano 6/fisiología , Herpesvirus Humano 7/genética , Herpesvirus Humano 7/inmunología , Herpesvirus Humano 7/patogenicidad , Herpesvirus Humano 7/fisiología , Humanos , Inmunidad Celular , Inmunidad Humoral , Lactante , Proteína Cofactora de Membrana/fisiología , Receptores Virales/fisiología , Linfocitos T/virología , Virión/patogenicidad , Activación Viral , Integración Viral , Latencia del Virus
15.
Int J Surg ; 83: 184-188, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32987208

RESUMEN

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.


Asunto(s)
Activación de Complemento , Xenoinjertos/inmunología , Proteína Cofactora de Membrana/fisiología , Animales , Animales Modificados Genéticamente , Aorta/inmunología , Citotoxicidad Inmunológica , Células Endoteliales/inmunología , Humanos , Porcinos
16.
Oncology ; 77(6): 366-77, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20068366

RESUMEN

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.


Asunto(s)
Adenocarcinoma/terapia , Adenoviridae/genética , Terapia Genética , Neoplasias Pulmonares/terapia , Proteína Cofactora de Membrana/fisiología , Receptores Virales/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Línea Celular Tumoral , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Femenino , Vectores Genéticos , Humanos , Masculino , Persona de Mediana Edad
17.
Front Immunol ; 10: 1074, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31164885

RESUMEN

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.


Asunto(s)
Antígenos CD55/fisiología , Antígenos CD59/fisiología , Inmunoterapia/métodos , Proteína Cofactora de Membrana/fisiología , Neoplasias/inmunología , Proteínas del Sistema Complemento/fisiología , Humanos , Neoplasias/etiología , Neoplasias/terapia , Receptores de Complemento 3b/fisiología
18.
Mol Immunol ; 44(1-3): 111-22, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16882452

RESUMEN

The hemolytic uremic syndrome is characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia and acute renal failure. There are two general types. One occurs in epidemic form and is diarrheal associated (D+HUS). It has a good prognosis. The second is a rare form known as atypical (aHUS), which may be familial or sporadic, and has a poor prognosis. aHUS is increasingly recognized to be a disease of defective complement regulation, particularly cofactor activity. Mutations in membrane cofactor protein (MCP; CD46) that predispose to the development of aHUS were first identified in 2003. MCP is a membrane-bound complement regulator that acts as a cofactor for the factor I-mediated cleavage of C3b and C4b deposited on host cells. More than 20 different mutations in MCP have now been identified in patients with aHUS. Many of these mutants have been functionally characterized and have helped to define the pathogenic mechanisms leading to aHUS development. Over 75% of the reported mutations cause a reduction in MCP expression, due to homozygous, compound heterozygous or heterozygous mutations. This deficiency of MCP leads to inadequate control of complement activation on endothelial cells after an initiating injury. The remaining MCP mutants are expressed, but demonstrate reduced ligand (C3b/C4b) binding capacity and cofactor activity of MCP. MCP mutations in aHUS demonstrate incomplete penetrance, indicating that additional genetic and environmental factors are required to manifest disease. MCP mutants as a cause of aHUS have a favorable clinical outcome in comparison to patients with factor H (CFH) or factor I (IF) mutations. In 90% of the renal transplants performed in patients with MCP-HUS, there has been no recurrence of the primary disease, whilst >50% of factor I or factor H deficient patients have had a prompt recurrence. This highlights the importance of defining and characterizing the underlying genetic defects in patients with aHUS.


Asunto(s)
Síndrome Hemolítico-Urémico/etiología , Proteína Cofactora de Membrana/genética , Mutación , Haplotipos , Síndrome Hemolítico-Urémico/clasificación , Síndrome Hemolítico-Urémico/genética , Humanos , Trasplante de Riñón , Proteína Cofactora de Membrana/química , Proteína Cofactora de Membrana/fisiología
19.
J Neuroimmunol ; 191(1-2): 70-8, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17936368

RESUMEN

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). In the recent years, accumulating evidence has supported an immunosuppressive role for regulatory T cells (Tregs). Most studies in the context of autoimmunity have focused on the defects of the CD4+CD25 high Tregs. However, we recently demonstrated an altered function of Tr1 Treg cells in MS, characterized by a lack of IL-10 secretion. Therefore, several major regulatory T cell defects are involved in human autoimmune disease. Hence, the induction of Tregs or the stimulation of Treg activity may be beneficial for the treatment of such diseases.


Asunto(s)
Proteína Cofactora de Membrana/fisiología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/fisiología , Animales , Diferenciación Celular , Herpesvirus Humano 6/fisiología , Humanos , Interleucina-10/biosíntesis , Activación de Linfocitos , Isoformas de Proteínas , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA