Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Med Microbiol Immunol ; 208(3-4): 475-485, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31065796

RESUMEN

Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects, yet there are no established treatments for preventing maternal-fetal transmission. During first trimester, HCMV replicates in basal decidua that functions as a reservoir for virus and source of transmission to the attached placenta and fetal hemiallograft but also contains immune cells, including natural killer cells, macrophages, and T cell subsets, that respond to pathogens, protecting the placenta and fetus. However, the specific cellular and cytokine responses to infection are unknown, nor are the immune correlates of protection that guide development of therapeutic strategies. Here we survey immune cell phenotypes in intact explants of basal decidua infected with a clinical pathogenic HCMV strain ex vivo and identify specific changes occurring in response to infection in the tissue environment. Using 4-color immunofluorescence microscopy, we found that at 3 days postinfection, virus replicates in decidual stromal cells and epithelial cells of endometrial glands. Infected cells and effector memory CD8+ T cells (TEM) in contact with them make IFN-γ. CD8+ TEM cells produce granulysin and cluster at sites of infection in decidua and the epithelium of endometrial glands. Quantification indicated expansion of two immune cell subtypes-CD8+ TEM cells and, to a lesser extent, iNKT cells. Approximately 20% of immune cells were found in pairs in both control and infected decidua, suggesting frequent cross-talk in the microenvironment of decidua. Our findings indicate a complex immune microenvironment in basal decidua and suggest CD8+ TEM cells play a role in early responses to decidual infection in seropositive women.


Asunto(s)
Infecciones por Citomegalovirus/patología , Citomegalovirus/crecimiento & desarrollo , Citomegalovirus/inmunología , Decidua/patología , Inmunidad Celular , Placenta/patología , Linfocitos T CD8-positivos/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Humanos , Células T Asesinas Naturales/inmunología , Técnicas de Cultivo de Órganos , Embarazo , Células del Estroma/patología , Células del Estroma/virología
2.
Am J Pathol ; 186(11): 2970-2986, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27638253

RESUMEN

Human cytomegalovirus (HCMV) is the leading viral cause of birth defects, including microcephaly, neurological deficits, hearing impairment, and vision loss. We previously reported that epithelial cells in amniotic membranes of placentas from newborns with intrauterine growth restriction and underlying congenital HCMV infection contain viral proteins in cytoplasmic vesicles. Herein, we immunostained amniotic membranes from 51 placentas from symptomatic and asymptomatic congenital infection with HCMV DNA in amniotic fluid and/or newborn saliva, intrauterine growth restriction, preterm deliveries, and controls. We consistently observed HCMV proteins in amniotic epithelial cells (AmEpCs) from infected placentas, sometimes with aberrant morphology. Primary AmEpCs isolated from mid-gestation placentas infected with pathogenic VR1814 proliferated and released infectious progeny for weeks, producing higher virus titers than late-gestation cells that varied by donor. In contrast to intact virion assembly compartments in differentiated retinal pigment epithelial cells, infected AmEpCs made dispersed multivesicular bodies. Primary AmEpCs and explants of amniochorionic membranes from mid-gestation placentas formed foci of infection, and interferon-ß production was prolonged. Infected AmEpCs up-regulated anti-apoptotic proteins survivin and Bcl-xL by mechanisms dependent and independent of the activated STAT3. Amniotic membranes naturally expressed both survivin and Bcl-xL, indicating that fetal membranes could foster persistent viral infection. Our results suggest strengthening innate immune responses and reducing viral functions could suppress HCMV infection in the fetal compartment.


Asunto(s)
Infecciones por Citomegalovirus/congénito , Citomegalovirus/inmunología , Placenta/virología , Complicaciones Infecciosas del Embarazo/virología , Amnios/patología , Amnios/virología , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Femenino , Retardo del Crecimiento Fetal/virología , Feto/metabolismo , Edad Gestacional , Humanos , Recién Nacido , Interferón beta/metabolismo , Placenta/patología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/patología , Carga Viral , Replicación Viral
3.
J Virol ; 89(9): 5134-47, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25741001

RESUMEN

UNLABELLED: Human cytomegalovirus (HCMV) is a major cause of birth defects that include severe neurological deficits, hearing and vision loss, and intrauterine growth restriction. Viral infection of the placenta leads to development of avascular villi, edema, and hypoxia associated with symptomatic congenital infection. Studies of primary cytotrophoblasts (CTBs) revealed that HCMV infection impedes terminal stages of differentiation and invasion by various molecular mechanisms. We recently discovered that HCMV arrests earlier stages involving development of human trophoblast progenitor cells (TBPCs), which give rise to the mature cell types of chorionic villi-syncytiotrophoblasts on the surfaces of floating villi and invasive CTBs that remodel the uterine vasculature. Here, we show that viral proteins are present in TBPCs of the chorion in cases of symptomatic congenital infection. In vitro studies revealed that HCMV replicates in continuously self-renewing TBPC lines derived from the chorion and alters expression and subcellular localization of proteins required for cell cycle progression, pluripotency, and early differentiation. In addition, treatment with a human monoclonal antibody to HCMV glycoprotein B rescues differentiation capacity, and thus, TBPCs have potential utility for evaluation of the efficacies of novel antiviral antibodies in protecting and restoring placental development. Our results suggest that HCMV replicates in TBPCs in the chorion in vivo, interfering with the earliest steps in the growth of new villi, contributing to virus transmission and impairing compensatory development. In cases of congenital infection, reduced responsiveness of the placenta to hypoxia limits the transport of substances from maternal blood and contributes to fetal growth restriction. IMPORTANCE: Human cytomegalovirus (HCMV) is a leading cause of birth defects in the United States. Congenital infection can result in permanent neurological defects, mental retardation, hearing loss, visual impairment, and pregnancy complications, including intrauterine growth restriction, preterm delivery, and stillbirth. Currently, there is neither a vaccine nor any approved treatment for congenital HCMV infection during gestation. The molecular mechanisms underlying structural deficiencies in the placenta that undermine fetal development are poorly understood. Here we report that HCMV replicates in trophoblast progenitor cells (TBPCs)-precursors of the mature placental cells, syncytiotrophoblasts and cytotrophoblasts, in chorionic villi-in clinical cases of congenital infection. Virus replication in TBPCs in vitro dysregulates key proteins required for self-renewal and differentiation and inhibits normal division and development into mature placental cells. Our findings provide insights into the underlying molecular mechanisms by which HCMV replication interferes with placental maturation and transport functions.


Asunto(s)
Diferenciación Celular , Infecciones por Citomegalovirus/patología , Citomegalovirus/fisiología , Placenta/virología , Células Madre/virología , Trofoblastos/virología , Replicación Viral , Infecciones por Citomegalovirus/virología , Femenino , Humanos , Embarazo , Complicaciones Infecciosas del Embarazo/patología , Complicaciones Infecciosas del Embarazo/virología , Células Madre/fisiología , Trofoblastos/fisiología
4.
Antimicrob Agents Chemother ; 59(3): 1558-68, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534746

RESUMEN

Human cytomegalovirus (HCMV) is the most common infection causing poor outcomes among transplant recipients. Maternal infection and transplacental transmission are major causes of permanent birth defects. Although no active vaccines to prevent HCMV infection have been approved, passive immunization with HCMV-specific immunoglobulin has shown promise in the treatment of both transplant and congenital indications. Antibodies targeting the viral glycoprotein B (gB) surface protein are known to neutralize HCMV infectivity, with high-affinity binding being a desirable trait, both to compete with low-affinity antibodies that promote the transmission of virus across the placenta and to displace nonneutralizing antibodies binding nearby epitopes. Using a miniaturized screening technology to characterize secreted IgG from single human B lymphocytes, 30 antibodies directed against gB were previously cloned. The most potent clone, TRL345, is described here. Its measured affinity was 1 pM for the highly conserved site I of the AD-2 epitope of gB. Strain-independent neutralization was confirmed for 15 primary HCMV clinical isolates. TRL345 prevented HCMV infection of placental fibroblasts, smooth muscle cells, endothelial cells, and epithelial cells, and it inhibited postinfection HCMV spread in epithelial cells. The potential utility for preventing congenital transmission is supported by the blockage of HCMV infection of placental cell types central to virus transmission to the fetus, including differentiating cytotrophoblasts, trophoblast progenitor cells, and placental fibroblasts. Further, TRL345 was effective at controlling an ex vivo infection of human placental anchoring villi. TRL345 has been utilized on a commercial scale and is a candidate for clinical evaluation.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Linfocitos B/virología , Línea Celular , Infecciones por Citomegalovirus/virología , Células Endoteliales/inmunología , Células Endoteliales/virología , Células Epiteliales/inmunología , Células Epiteliales/virología , Epítopos/inmunología , Femenino , Fibroblastos/inmunología , Fibroblastos/virología , Humanos , Inmunoglobulina G/inmunología , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/virología , Placenta/inmunología , Placenta/virología , Embarazo , Proteínas del Envoltorio Viral/inmunología
5.
J Infect Dis ; 209(10): 1573-84, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24403553

RESUMEN

BACKGROUND: Human cytomegalovirus (HCMV) is the major viral etiology of congenital infection and birth defects. Fetal transmission is high (30%-40%) in primary maternal infection, and symptomatic babies have permanent neurological, hearing, and vision defects. Recurrent infection is infrequently transmitted (2%) and largely asymptomatic. Congenital infection is also associated with intrauterine growth restriction (IUGR). METHODS: To investigate possible underlying HCMV infection in cases of idiopathic IUGR, we studied maternal and cord sera and placentas from 19 pregnancies. Anti-HCMV antibodies, hypoxia-related factors, and cmvIL-10 were measured in sera. Placental biopsy specimens were examined for viral DNA, expression of infected cell proteins, and pathology. RESULTS: Among 7 IUGR cases, we identified 2 primary and 3 recurrent HCMV infections. Virus replicated in glandular epithelium and lymphatic endothelium in the decidua, cytotrophoblasts, and smooth muscle cells in blood vessels of floating villi and the chorion. Large fibrinoids with avascular villi, edema, and inflammation were significantly increased. Detection of viral proteins in the amniotic epithelium indicated transmission in 2 cases of IUGR with primary infection and 3 asymptomatic recurrent infections. CONCLUSIONS: Congenital HCMV infection impairs placental development and functions and should be considered as an underlying cause of IUGR, regardless of virus transmission to the fetus.


Asunto(s)
Infecciones por Citomegalovirus/complicaciones , Retardo del Crecimiento Fetal/virología , Complicaciones Infecciosas del Embarazo/patología , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , ADN Viral , Femenino , Humanos , Inmunoglobulina G/sangre , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa , Proyectos Piloto , Embarazo , Pruebas Serológicas
6.
Am J Pathol ; 181(5): 1540-59, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22959908

RESUMEN

We investigated human cytomegalovirus pathogenesis by comparing infection with the low-passage, endotheliotropic strain VR1814 and the attenuated laboratory strain AD169 in human placental villi as explants in vitro and xenografts transplanted into kidney capsules of SCID mice (ie, mice with severe combined immunodeficiency). In this in vivo human placentation model, human cytotrophoblasts invade the renal parenchyma, remodel resident arteries, and induce a robust lymphangiogenic response. VR1814 replicated in villous and cell column cytotrophoblasts and reduced formation of anchoring villi in vitro. In xenografts, infected cytotrophoblasts had a severely diminished capacity to invade and remodel resident arteries. Infiltrating lymphatic endothelial cells proliferated, aggregated, and failed to form lymphatic vessels. In contrast, AD169 grew poorly in cytotrophoblasts in explants, and anchoring villi formed normally in vitro. Likewise, viral replication was impaired in xenografts, and cytotrophoblasts retained invasive capacity, but some partially remodeled blood vessels incorporated lymphatic endothelial cells and were permeable to blood. The expression of both vascular endothelial growth factor (VEGF)-C and basic fibroblast growth factor increased in VR1814-infected explants, whereas VEGF-A and soluble VEGF receptor-3 increased in those infected with AD169. Our results suggest that viral replication and paracrine factors could undermine vascular remodeling and cytotrophoblast-induced lymphangiogenesis, contributing to bleeding, hypoxia, and edema in pregnancies complicated by congenital human cytomegalovirus infection.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Infecciones por Citomegalovirus/patología , Citomegalovirus/fisiología , Linfangiogénesis , Placentación , Trofoblastos/patología , Trofoblastos/virología , Animales , Arterias/patología , Vasos Sanguíneos/patología , Movimiento Celular , Proliferación Celular , Vellosidades Coriónicas/crecimiento & desarrollo , Vellosidades Coriónicas/patología , Vellosidades Coriónicas/trasplante , Vellosidades Coriónicas/virología , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/fisiopatología , Infecciones por Citomegalovirus/virología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Riñón/irrigación sanguínea , Riñón/patología , Ratones , Ratones SCID , Embarazo , Células Madre/patología , Células Madre/virología , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Replicación Viral
7.
Vaccines (Basel) ; 7(4)2019 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-31569508

RESUMEN

Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects worldwide, yet the most effective strategies for preventing virus transmission during pregnancy are unknown. We measured the efficacy of human monoclonal antibodies (mAbs) to HCMV attachment/entry factors glycoprotein B (gB) and the pentameric complex, gH/gL-pUL128-131, in preventing infection and spread of a clinical strain in primary placental cells and explants of developing anchoring villi. A total of 109 explants from five first-trimester placentas were cultured, and infection was analyzed in over 400 cell columns containing ~120,000 cytotrophoblasts (CTBs). mAbs to gB and gH/gL, 3-25 and 3-16, respectively, neutralized infection in stromal fibroblasts and trophoblast progenitor cells. mAbs to pUL128-131 of the pentameric complex, 1-103 and 2-18, neutralized infection of amniotic epithelial cells better than mAbs 3-25 and 3-16 and hyperimmune globulin. Select mAbs neutralized infection of cell column CTBs, with mAb 2-18 most effective, followed by mAb 3-25. Treatment of anchoring villi with mAbs postinfection reduced spread in CTBs and impaired formation of virion assembly compartments, with mAb 2-18 achieving better suppression at lower concentrations. These results predict that antibodies generated by HCMV vaccines or used for passive immunization have the potential to reduce transplacental transmission and congenital disease.

8.
Placenta ; 59 Suppl 1: S8-S16, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28477968

RESUMEN

Congenital human cytomegalovirus (HCMV) infection is a major viral cause of birth defects, including microcephaly, neurological deficits, loss of hearing and vision, and intrauterine growth restriction. Despite its public health significance, there is no approved treatment for congenital infection during pregnancy; existing antivirals have unacceptable toxicities. The mechanisms of HCMV-induced placental injury, reduced capacity for compensatory development and transmission to the fetus are poorly understood, limiting the development of alternative strategies for clinical management of the disease. Recently, self-renewing, multipotent trophoblast progenitor cells (TBPCs) were reported to reside in the chorion of the human placenta and differentiate into the mature trophoblast subtypes - transport syncytiotrophoblasts and invasive cytotrophoblasts - forming chorionic villi, the functional units of the placenta. HCMV infects TBPCs, reducing the population of progenitor cells and their functional capacity to self-renew, migrate and differentiate. Human TBPCs and chorionic villus explants from first trimester represent relevant models for evaluating efficacies of new antiviral agents in protecting and restoring growth of the developing placenta in response to adverse conditions. Correlating pathology from complications of congenital HCMV infection with impaired development in the tissue environment of anchoring villus explants and defects in TBPC differentiation may enable identification of molecular pathways that could serve as targets for intervention. Here we summarize studies that could open up novel avenues of research on potential therapeutics to sustain placental development, promote differentiation and improve function and pregnancy outcomes.


Asunto(s)
Infecciones por Citomegalovirus/fisiopatología , Placentación , Complicaciones del Embarazo/fisiopatología , Anticuerpos Monoclonales/uso terapéutico , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/terapia , Femenino , Humanos , Embarazo , Trofoblastos/virología , Proteínas del Envoltorio Viral/inmunología
9.
Cell Host Microbe ; 20(2): 155-66, 2016 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-27443522

RESUMEN

Zika virus (ZIKV) infection during pregnancy is linked to severe birth defects, but mother-to-fetus transmission routes are unknown. We infected different primary cell types from mid- and late-gestation placentas and explants from first-trimester chorionic villi with the prototype Ugandan and a recently isolated Nicaraguan ZIKV strain. ZIKV infects primary human placental cells and explants-cytotrophoblasts, endothelial cells, fibroblasts, and Hofbauer cells in chorionic villi and amniotic epithelial cells and trophoblast progenitors in amniochorionic membranes-that express Axl, Tyro3, and/or TIM1 viral entry cofactors. ZIKV produced NS3 and E proteins and generated higher viral titers in amniotic epithelial cells from mid-gestation compared to late-gestation placentas. Duramycin, a peptide that binds phosphatidylethanolamine in enveloped virions and precludes TIM1 binding, reduced ZIKV infection in placental cells and explants. Our results suggest that ZIKV spreads from basal and parietal decidua to chorionic villi and amniochorionic membranes and that targeting TIM1 could suppress infection at the uterine-placental interface.


Asunto(s)
Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Transmisión Vertical de Enfermedad Infecciosa , Placenta/virología , Receptores Virales/metabolismo , Tropismo Viral , Infección por el Virus Zika/transmisión , Virus Zika/fisiología , Antivirales/metabolismo , Bacteriocinas/metabolismo , Células Cultivadas , Femenino , Humanos , Técnicas de Cultivo de Órganos , Péptidos/metabolismo , Embarazo , Internalización del Virus/efectos de los fármacos , Infección por el Virus Zika/virología
10.
Viruses ; 6(3): 1346-64, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24651029

RESUMEN

Human cytomegalovirus (HCMV) is the major viral cause of congenital infection and birth defects. Primary maternal infection often results in virus transmission, and symptomatic babies can have permanent neurological deficiencies and deafness. Congenital infection can also lead to intrauterine growth restriction, a defect in placental transport. HCMV replicates in primary cytotrophoblasts (CTBs), the specialized cells of the placenta, and inhibits differentiation/invasion. Human trophoblast progenitor cells (TBPCs) give rise to the mature cell types of the chorionic villi, CTBs and multi-nucleated syncytiotrophoblasts (STBs). Here we report that TBPCs are fully permissive for pathogenic and attenuated HCMV strains. Studies with a mutant virus lacking a functional pentamer complex (gH/gL/pUL128-131A) showed that virion entry into TBPCs is independent of the pentamer. In addition, infection is blocked by a potent human neutralizing monoclonal antibody (mAb), TRL345, reactive with glycoprotein B (gB), but not mAbs to the pentamer proteins pUL130/pUL131A. Functional studies revealed that neutralization of infection preserved the capacity of TBPCs to differentiate and assemble into trophospheres composed of CTBs and STBs in vitro. Our results indicate that mAbs to gB protect trophoblast progenitors of the placenta and could be included in antibody treatments developed to suppress congenital infection and prevent disease.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Citomegalovirus/inmunología , Células Madre/virología , Trofoblastos/virología , Proteínas del Envoltorio Viral/inmunología , Células Cultivadas , Citomegalovirus/fisiología , Femenino , Humanos , Placenta/inmunología , Embarazo , Tropismo Viral , Internalización del Virus
11.
J Clin Virol ; 46 Suppl 4: S58-63, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19854676

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) is the major cause of congenital infection and disease leading to permanent birth defects. In about 35-40% of pregnant women with primary CMV infection, virus crosses the placenta, resulting in the birth of congenitally infected babies. In contrast, this happens in only 1-3% of seropositive women with strong CMV-specific humoral immunity. Whether CMV reaches the fetus and disseminates depends on the level of high-avidity antibodies in the maternal circulation and the passive immunity of the fetus. OBJECTIVES AND STUDY DESIGN: To identify CMV infection in uncomplicated deliveries based on detection of viral DNA in placental biopsy specimens at term. To quantify CMV-specific IgG avidity, neutralizing titer, IgG1 concentration, and characterize the immunoblot profiles for CMV proteins in paired samples of placental and cord blood sera. RESULTS: In accord with earlier reports, CMV DNA was detected in 39% (11/28) of placentas with mean- to high-avidity CMV-specific IgG. In seropositive women, the concentration of antiviral antibodies, specifically IgG1, increased in the fetal bloodstream, and CMV neutralizing titers in maternal and fetal blood were comparable. CONCLUSIONS: CMV-specific, high-avidity neutralizing antibodies from maternal circulation are transcytosed to the fetal bloodstream, contribute to suppression of viral replication in the placenta and could prevent congenital disease.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Feto/inmunología , Inmunoglobulina G/sangre , Complicaciones Infecciosas del Embarazo/inmunología , Afinidad de Anticuerpos/inmunología , Infecciones por Citomegalovirus/sangre , Femenino , Sangre Fetal/inmunología , Sangre Fetal/virología , Feto/virología , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Pruebas de Neutralización , Placenta/inmunología , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/sangre , Complicaciones Infecciosas del Embarazo/virología
12.
Am J Pathol ; 172(4): 1127-40, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18349127

RESUMEN

Human cytomegalovirus (CMV) infection is a major cause of morbidity in immunosuppressed individuals, and congenital CMV infection is a leading cause of birth defects in newborns. Infection with pathogenic viral strains alters cell-cell and cell-matrix interactions, affecting extracellular matrix remodeling and endothelial cell migration. The multifunctional cytokine transforming growth factor (TGF)-beta1 regulates cell proliferation, differentiation, and extracellular matrix remodeling. Secreted as a latent protein complex, TGF-beta1 requires activation before binding to receptors that phosphorylate intracellular effectors. TGF-beta1 is activated by integrin alphavbeta6, which is strongly induced in the epithelium by injury and inflammation but has not previously been found in endothelial cells. Here, we report that CMV infection induces integrin alphavbeta6 expression in endothelial cells, leading to activation of TGF-beta1, signaling through its receptor ALK5, and phosphorylation of its intracellular effector Smad3. Infection of endothelial cells was also found to stimulate collagen synthesis through a mechanism dependent on both TGF-beta1 and integrin alphavbeta6. Immunohistochemical analysis showed integrin alphavbeta6 up-regulation in capillaries proximal to foci of CMV infection in lungs, salivary glands, uterine decidua, and injured chorionic villi of the placenta, demonstrating both its induction in endothelium and up-regulation in epithelium in vivo. Our results suggest that activation of TGF-beta1 by integrin alphavbeta6 contributes to pathological changes and may impair endothelial cell functions in tissues that are chronically infected with CMV.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Colágeno/biosíntesis , Infecciones por Citomegalovirus/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/virología , Células Epiteliales/metabolismo , Integrinas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Receptores de Activinas Tipo II/metabolismo , Animales , Células Cultivadas , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/virología , Células Endoteliales/patología , Matriz Extracelular/metabolismo , Humanos , Cadenas beta de Integrinas/metabolismo , Visón , Especificidad de Órganos , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Venas Umbilicales/patología , Venas Umbilicales/virología , Regulación hacia Arriba , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA