Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
HIV Med ; 25(6): 692-699, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38350653

RESUMEN

OBJECTIVES: In countries with access to early antiretroviral treatment (ART), opportunistic infections caused by cytomegalovirus (CMV) in people living with HIV (PLWH) are becoming increasingly rare. As potential complications are severe, it is critical to remain aware of this important diagnosis. However, clinical characteristics and prognosis of CMV infection in PLWH in the era of modern ART have not been well described. METHODS: Here, we compiled the clinical presentation, management and outcome of CMV infection in PLWH treated at the infectious diseases clinic of Karolinska University Hospital during 2010-2020. RESULTS: We identified 51 cases of active CMV infection, based on detection of CMV-DNA, mainly diagnosed in patients with CD4 T-cell count <200 cells/µL (86%). Median time from HIV diagnosis to detection of CMV infection was 16 days. In 20 cases (39%), CMV infection was symptomatic with retinitis identified as a manifestation in 70% of cases. Symptomatic CMV infection was treated for 73 (20-313) days upon diagnosis, mostly using valganciclovir. One-year mortality was 22% and was associated with longer time to ART initiation from HIV diagnosis and with comorbidities, but not with CMV-DNA levels or CD4 count. Immune reconstitution was not significantly compromised in patients with symptomatic CMV, although CD4/8 ratio tended to be lower in patients with systemic CMV infection. CONCLUSIONS: Retinitis remains the most common manifestation of symptomatic CMV infection in PLWH. Recognizing CMV infection is important, especially in the management of 'late presenters'. Adequate duration of antiviral therapy and appropriate follow-up must be ensured to avoid complications.


Asunto(s)
Antivirales , Infecciones por Citomegalovirus , Infecciones por VIH , Hospitales Universitarios , Humanos , Masculino , Femenino , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/complicaciones , Adulto , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/complicaciones , Persona de Mediana Edad , Suecia/epidemiología , Recuento de Linfocito CD4 , Antivirales/uso terapéutico , Infecciones Oportunistas Relacionadas con el SIDA/tratamiento farmacológico , Infecciones Oportunistas Relacionadas con el SIDA/diagnóstico , Resultado del Tratamiento , Citomegalovirus/aislamiento & purificación , Valganciclovir/uso terapéutico
2.
Medicina (Kaunas) ; 59(7)2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37512060

RESUMEN

Background: Human cytomegalovirus (HCMV) has been detected in tissue samples from patients with glioblastoma but little is known about the systemic immunological response to HCMV in these patients. Objectives: To investigate the presence and clinical significance of HCMV antibodies levels in plasma samples obtained from patients with brain tumors. Materials and Methods: HCMV-specific IgG and IgM antibody levels were determined in 59 plasma samples collected from brain tumor patients included in a prospective study and in 114 healthy individuals. We examined if the levels of HCMV specific antibodies varied in patients with different brain tumor diagnoses compared to healthy individuals, and if antibody levels were predictive for survival time. Results: HCMV specific IgG antibodies were detected by ELISA in 80% and 89% of patients with GBM and astrocytoma grades II-III, respectively, in all samples (100%) from patients with secondary GBM and brain metastases, as well as in 80% of healthy donors (n = 114). All plasma samples were negative for HCMV-IgM. Patients with brain metastases who had higher plasma HCMV-IgG titers had longer survival times (p = 0.03). Conclusions: HCMV specific IgG titers were higher among all brain tumor patient groups compared with healthy donors, except for patients with secondary GBM. Higher HCMV specific IgG levels in patients with brain metastases but not in patients with primary brain tumors were associated with prolonged survival time.


Asunto(s)
Neoplasias Encefálicas , Infecciones por Citomegalovirus , Humanos , Citomegalovirus , Infecciones por Citomegalovirus/complicaciones , Estudios Prospectivos , Anticuerpos Antivirales , Inmunoglobulina G
3.
J Med Virol ; 93(6): 4023-4027, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33174621

RESUMEN

Borderline ovarian tumors (BOTs) belong to a group of tumors that are distinctly different from ovarian carcinomas. There is an increased risk of BOTs in patients with pelvic inflammatory disease. Human cytomegalovirus (HCMV) has been detected in ovarian cancer tissue specimens. This virus favors the inflammatory milieu by inducing expression of the potent inflammatory factor 5-lipoxygenase (5LO), which stimulates cellular viability, cellular proliferation and activates antiapoptotic signaling pathways. Here, we aimed to examine presence of HCMV and 5LO in BOTs. Expression levels of HCMV proteins (IE and pp65) and 5LO were examined in paraffin embedded BOT tissue sections by immunohistochemistry staining and HCMV immunoglobulin M and immunoglobulin G (IgG) levels were determined by serology in blood samples obtained from 15 patients with BOTs identified in a prospective study at Karolinska University Hospital. Extensive expression of HCMV-IE, pp65, and 5LO were detected in 87%, 40%, and 90% of examined BOT tissue sections, respectively. HCMV-IgG prevalence and antibody levels were significantly higher in patients with BOT compared to age matched healthy women (83.3% vs. 65,6%, respectively, p = .01). Whether HCMV can induce inflammation and affect the pathogenesis of BOTs should therefore be further investigated.


Asunto(s)
Araquidonato 5-Lipooxigenasa/genética , Infecciones por Citomegalovirus/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/virología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/sangre , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/virología , Femenino , Humanos , Inmunohistoquímica , Inflamación/genética , Persona de Mediana Edad , Neoplasias Ováricas/fisiopatología , Adhesión en Parafina , Estudios Prospectivos
4.
Immun Ageing ; 18(1): 12, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712035

RESUMEN

The majority of people infected with SARS-CoV-2 are asymptomatic or have mild to moderate symptoms. However, for unknown reasons, about 15 % have severe pneumonia requiring hospital care and oxygen support, and about 5 % develop acute respiratory distress syndrome, septic shock, and multiorgan failure that result in a high mortality rate. The risk of severe COVID-19 is highest among those who are over 70 years of age. Why severe COVID-19 develops in some people but not others is not understood. Could some cases involve reactivation of latent cytomegalovirus (CMV)?

5.
J Biol Chem ; 294(44): 16297-16308, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31519750

RESUMEN

Herpesviruses can rewire cellular signaling in host cells by expressing viral G protein-coupled receptors (GPCRs). These viral receptors exhibit homology to human chemokine receptors, but some display constitutive activity and promiscuous G protein coupling. Human cytomegalovirus (HCMV) has been detected in multiple cancers, including glioblastoma, and its genome encodes four GPCRs. One of these receptors, US28, is expressed in glioblastoma and possesses constitutive activity and oncomodulatory properties. UL33, another HCMV-encoded GPCR, also displays constitutive signaling via Gαq, Gαi, and Gαs proteins. However, little is known about the nature and functional effects of UL33-driven signaling. Here, we assessed UL33's signaling repertoire and oncomodulatory potential. UL33 activated multiple proliferative, angiogenic, and inflammatory signaling pathways in HEK293T and U251 glioblastoma cells. Notably, upon infection, UL33 contributed to HCMV-mediated STAT3 activation. Moreover, UL33 increased spheroid growth in vitro and accelerated tumor growth in different in vivo tumor models, including an orthotopic glioblastoma xenograft model. UL33-mediated signaling was similar to that stimulated by US28; however, UL33-induced tumor growth was delayed. Additionally, the spatiotemporal expression of the two receptors only partially overlapped in HCMV-infected glioblastoma cells. In conclusion, our results unveil that UL33 has broad signaling capacity and provide mechanistic insight into its functional effects. UL33, like US28, exhibits oncomodulatory properties, elicited via constitutive activation of multiple signaling pathways. UL33 and US28 might contribute to HCMV's oncomodulatory effects through complementing and converging cellular signaling, and hence UL33 may represent a promising drug target in HCMV-associated malignancies.


Asunto(s)
Receptores de Quimiocina/metabolismo , Proteínas Virales/metabolismo , Animales , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Citomegalovirus/metabolismo , Proteínas de Unión al GTP/metabolismo , Glioblastoma/patología , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Receptores de Quimiocina/genética , Receptores Virales/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
6.
Int J Cancer ; 142(6): 1266-1276, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29067692

RESUMEN

Glioma stem cells (GSCs) are glioblastoma (GBM) cells that are resistant to therapy and can give rise to recurrent tumors. The identification of patient-related factors that support GSCs is thus necessary to design effective therapies for GBM patients. Glucocorticoids (GCs) are used to treat GBM-associated edema. However, glucocorticoids participate in the physiological response to psychosocial stress, which has been linked to poor cancer prognosis. This raises concern that glucocorticoids affect the tumor and GSCs. Here, we treated primary human GBM cells with dexamethasone and evaluated GC-driven changes in cell morphology, proliferation, migration, gene expression, secretory activity and growth as neurospheres. Dexamethasone treatment of GBM cells appeared to promote the development of a GSC-like phenotype and conferred resistance to physiological stress and chemotherapy. We also analyzed a potential correlation between GC treatment and tumor recurrence after surgical excision in a population-based consecutive cohort of 48 GBM patients, adjusted for differences in known prognostic factors concerning baseline and treatment characteristics. In this cohort, we found a negative correlation between GC intake and progression-free survival, regardless of the MGMT methylation status. In conclusion, our findings raise concern that treatment of GBM with GCs may compromise the efficacy of chemotherapy and may support a GSC population, which could contribute to tumor recurrence and the poor prognosis of the disease.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/terapia , Resistencia a Antineoplásicos/efectos de los fármacos , Glioblastoma/terapia , Glucocorticoides/efectos adversos , Recurrencia Local de Neoplasia/prevención & control , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/uso terapéutico , Encéfalo/patología , Encéfalo/cirugía , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Quimioradioterapia Adyuvante/métodos , Dexametasona/efectos adversos , Supervivencia sin Enfermedad , Femenino , Glioblastoma/complicaciones , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Procedimientos Neuroquirúrgicos , Cultivo Primario de Células , Pronóstico , Estrés Fisiológico/efectos de los fármacos , Resultado del Tratamiento , Células Tumorales Cultivadas
7.
Breast Cancer Res Treat ; 172(2): 339-351, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30132219

RESUMEN

PURPOSE: We have compared the mutational profiles of human breast cancer tumor samples belonging to all major subgroups with special emphasis on triple-negative breast cancer (TNBC). Our major goal was to identify specific mutations that could be potentially used for clinical decision making in TNBC patients. PATIENTS AND METHODS: Primary tumor specimens from 149 Norwegian breast cancer patients were available. We analyzed the tissue samples for somatic mutations in 44 relevant breast cancer genes by targeted next-generation sequencing. As a second confirmatory technique, we performed pyrosequencing on selected samples. RESULTS: We observed a distinct subgroup of TNBC patients, characterized by an almost completely lack of pathogenic somatic mutations. A point mutation in the adenoviral E1A binding protein p300 (EP300-G211S) was significantly correlated to this TNBC subgroup. The EP300-G211S mutation was exclusively found in the TNBC patients and its presence reduced the chance for other pathological somatic mutations in typical breast cancer genes investigated in our gene panel by 94.9% (P < 0.005). Interestingly, the EP300-G211S mutation also predicted a lower risk for relapses and decreased breast cancer-specific mortality during long-term follow-up of the patients. CONCLUSION: Next-generation sequencing revealed specific mutations in EP300 to be associated with the mutational patterns in typical breast cancer genes and long-term outcome of triple-negative breast cancer patients.


Asunto(s)
Análisis Mutacional de ADN , Proteína p300 Asociada a E1A/genética , Recurrencia Local de Neoplasia/genética , Neoplasias de la Mama Triple Negativas/genética , Adulto , Anciano , Biomarcadores de Tumor/genética , Supervivencia sin Enfermedad , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Persona de Mediana Edad , Mutación , Recurrencia Local de Neoplasia/patología , Neoplasias de la Mama Triple Negativas/patología
8.
Acta Neuropathol ; 135(5): 779-798, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29305721

RESUMEN

Tumours can be viewed as aberrant tissues or organs sustained by tumorigenic stem-like cells that engage into dysregulated histo/organogenetic processes. Paragangliomas, prototypical organoid tumours constituted by dysmorphic variants of the vascular and neural tissues found in normal paraganglia, provide a model to test this hypothesis. To understand the origin of paragangliomas, we built a biobank comprising 77 cases, 18 primary cultures, 4 derived cell lines, 80 patient-derived xenografts and 11 cell-derived xenografts. We comparatively investigated these unique complementary materials using morphofunctional, ultrastructural and flow cytometric assays accompanied by microRNA studies. We found that paragangliomas contain stem-like cells with hybrid mesenchymal/vasculoneural phenotype, stabilized and expanded in the derived cultures. The viability and growth of such cultures depended on the downregulation of the miR-200 and miR-34 families, which allowed high PDGFRA and ZEB1 protein expression levels. Both tumour tissue- and cell culture-derived xenografts recapitulated the vasculoneural paraganglioma structure and arose from mesenchymal-like cells through a fixed developmental sequence. First, vasculoangiogenesis organized the microenvironment, building a perivascular niche which in turn supported neurogenesis. Neuroepithelial differentiation was associated with severe mitochondrial dysfunction, not present in cultured paraganglioma cells, but acquired in vivo during xenograft formation. Vasculogenesis was the Achilles' heel of xenograft development. In fact, imatinib, that targets endothelial-mural signalling, blocked paraganglioma xenograft formation (11 xenografts from 12 cell transplants in the control group versus 2 out of 10 in the treated group, P = 0.0015). Overall our key results were unaffected by the SDHx gene carrier status of the patient, characterized for 70 out of 77 cases. In conclusion, we explain the biphasic vasculoneural structure of paragangliomas and identify an early and pharmacologically actionable phase of paraganglioma organization.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/fisiopatología , Mesilato de Imatinib/uso terapéutico , Paraganglioma/tratamiento farmacológico , Paraganglioma/fisiopatología , Animales , Antineoplásicos/farmacología , Línea Celular , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Mesilato de Imatinib/farmacología , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Organogénesis/efectos de los fármacos , Organogénesis/fisiología , Paraganglioma/genética , Paraganglioma/patología , Cultivo Primario de Células , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
BMC Cancer ; 18(1): 154, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29409474

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the most common malignant brain tumor with median survival of 12-15 months. Owing to uncertainty in clinical outcome, additional prognostic marker(s) apart from existing markers are needed. Since overexpression of endothelin B receptor (ETBR) has been demonstrated in gliomas, we aimed to test whether ETBR is a useful prognostic marker in GBM and examine if the clinically available endothelin receptor antagonists (ERA) could be useful in the disease treatment. METHODS: Data from The Cancer Genome Atlas and the Gene Expression Omnibus database were analyzed to assess ETBR expression. For survival analysis, glioblastoma samples from 25 Swedish patients were immunostained for ETBR, and the findings were correlated with clinical history. The druggability of ETBR was assessed by protein-protein interaction network analysis. ERAs were analyzed for toxicity in in vitro assays with GBM and breast cancer cells. RESULTS: By bioinformatics analysis, ETBR was found to be upregulated in glioblastoma patients, and its expression levels were correlated with reduced survival. ETBR interacts with key proteins involved in cancer pathogenesis, suggesting it as a druggable target. In vitro viability assays showed that ERAs may hold promise to treat glioblastoma and breast cancer. CONCLUSIONS: ETBR is overexpressed in glioblastoma and other cancers and may be a prognostic marker in glioblastoma. ERAs may be useful for treating cancer patients.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Receptor de Endotelina B/genética , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Antagonistas de los Receptores de Endotelina/uso terapéutico , Femenino , Redes Reguladoras de Genes , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Pronóstico , Receptor de Endotelina B/metabolismo
11.
J Gen Virol ; 98(5): 1058-1072, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28589873

RESUMEN

Human cytomegalovirus (HCMV) infection results in the production of virions, dense bodies (DBs) and non-infectious enveloped particles, all of which incorporate proteins and RNAs that can be transferred to host cells. Here, we investigated whether virions and DBs also carry microRNAs (miRNAs) and assessed their delivery and functionality in cells. Human lung fibroblasts (MRC-5) were infected with the HCMV strain AD169, and conditioned cell culture medium was collected and centrifuged. The pellets were treated with RNase-ONE, and the virions and DBs were purified with a potassium tartrate-glycerol gradient and dialysed. The virions and DBs were incubated with micrococcal nuclease, DNA and RNA were extracted and then analysed with TaqMan PCR assays, while the proteins were examined with Western blots. To assess the delivery of miRNAs to cells and their functionality, virions and DBs were irradiated with UV light. The purity of the virions and DBs was confirmed by typical morphology, the presence of the structural protein pp65 and the HCMV genome, the ability to infect MRC-5 cells and the absence of the host genome. RNA analysis revealed the presence of 14 HCMV-encoded miRNAs (UL22A-5p, US25-1-5p, UL22A-3p, US5-2-3p, UL112-3p, US25-2-3p, US25-2-5p, US33-3p, US5-1, UL36-5p, US4-5p, UL36-3p, UL70-5p and US25-1-3p), HCMV immediate-early mRNA and long non-coding RNA2.7, moreover, two host-encoded miRNAs (hsa-miR-218-5p and hsa-miR-21-5p) and beta-2-microglobulin RNA. UV-irradiated virions and DBs delivered viral miRNAs (US25-1-5p and UL112-3p) to the host cells, and miR-US25-1-5p was functional in a luciferase reporter assay. We conclude that virions and DBs carry miRNAs that are biologically functional and can be delivered to cells, which may affect cellular processes.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/metabolismo , MicroARNs/metabolismo , ARN Viral/metabolismo , Virión/metabolismo , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Interacciones Huésped-Patógeno , Humanos , MicroARNs/genética , ARN Viral/genética , Virión/genética
12.
J Pediatr Gastroenterol Nutr ; 64(1): 76-82, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27203397

RESUMEN

OBJECTIVES: Human cytomegalovirus (HCMV) infection induces production of CD13-specific autoantibodies, which may promote inflammation and tissue damage. HCMV infection has been suggested as a cause of biliary atresia (BA), but little is known of its role in other forms of neonatal cholestasis. We studied serum levels of CD13-specific autoantibodies in mothers of infants with neonatal cholestasis of different causes, including BA, and in mothers of healthy, term infants without cholestasis, as well as in healthy blood donors. METHODS: Using fluorescence-activated cell sorting, we measured CD13-specific autoantibody levels in serum from the above-mentioned groups. In addition, the effect of serum from mothers of infants with neonatal cholestasis was tested on the differentiation of monocytes into macrophages. RESULTS: CD13-specific autoantibodies were found in mothers of infants with neonatal cholestasis, but not in mothers of infants without cholestasis and healthy blood donors, and were associated with HCMV seropositivity. Sera from mothers of infants with all forms of neonatal cholestasis inhibited differentiation of monocytes into macrophages, but this was not dependent on CD13-specific autoantibodies. CONCLUSIONS: The significantly higher frequency of CD13-specific autoantibodies in mothers of infants with neonatal cholestasis of all forms compared with mothers of healthy infants without cholestasis suggests an association, but does not prove that they are pathogenic. The presence of CD13-specific autoantibodies does not correlate with HCMV IgG serostatus, suggesting a more complicated mechanism that possibly reflects active HCMV infection in these individuals. Further studies are needed to elucidate whether these autoantibodies contribute to the development of cholestasis or represent an epiphenomenon.


Asunto(s)
Autoanticuerpos/sangre , Antígenos CD13 , Colestasis/sangre , Infecciones por Citomegalovirus/sangre , Citomegalovirus , Enfermedades del Recién Nacido/sangre , Complicaciones Infecciosas del Embarazo , Adulto , Animales , Atresia Biliar/sangre , Atresia Biliar/etiología , Colestasis/etiología , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/virología , Femenino , Humanos , Inmunoglobulina G/sangre , Recién Nacido , Enfermedades del Recién Nacido/etiología , Hepatopatías/sangre , Hepatopatías/etiología , Ratones , Monocitos , Madres , Células 3T3 NIH , Embarazo , Complicaciones Infecciosas del Embarazo/sangre , Complicaciones Infecciosas del Embarazo/virología , Adulto Joven
14.
J Gen Virol ; 96(12): 3598-3612, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26416316

RESUMEN

Human cytomegalovirus (hCMV) is a beta herpesvirus that establishes lifelong infection. Although the virus does not usually cause overt clinical symptoms in immunocompetent individuals it can have deleterious effects in immunocompromised patients, such as those on post-transplant medication or with HIV infection. hCMV is the most common congenital infection and can lead to serious fetal sequelae. Endothelial cells (ECs) are natural hosts for hCMV in vivo, therefore, investigations of how this cell type is modulated by infection are key to understanding hCMV pathogenesis. Previous studies have examined the effect of secretomes from hCMV-infected cells on EC angiogenesis, whereas the effect of direct infection on this process has not been so well investigated. Here, we show that placental ECs are viral targets during congenital infection and that vessels in infected tissue appear morphologically abnormal. We demonstrate that the clinical hCMV strain VR1814 impaired EC tube assembly in in vitro angiogenesis assays and inhibited wound healing ability in scratch assays. Secretomes from infected cultures did not impair angiogenesis of uninfected ECs, suggesting that cell-intrinsic changes, as opposed to secreted factors, were responsible. We observed viral gene transcription dependent downregulation of the expression of angiogenesis-associated genes, including angiopoietin-2, TEK receptor and vascular endothelial growth factor receptors. An alternative clinical hCMV stain, TB40E showed similar effects on EC angiogenesis. Together, our data indicate that direct infection with hCMV can induce an anti-migratory and anti-angiogenic EC phenotype, which could have a detrimental effect on the vasculature development in infected tissues.


Asunto(s)
Movimiento Celular , Infecciones por Citomegalovirus/congénito , Citomegalovirus/fisiología , Células Endoteliales/fisiología , Células Endoteliales/virología , Neovascularización Fisiológica , Células Cultivadas , Infecciones por Citomegalovirus/virología , Femenino , Regulación Viral de la Expresión Génica , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Interleucina-10/genética , Interleucina-10/metabolismo , Placenta/irrigación sanguínea , Placenta/citología , Placenta/virología , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 34(3): 571-80, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371083

RESUMEN

OBJECTIVE: A growing body of evidence indicates that platelets contribute to the onset and progression of atherosclerosis by modulating immune responses. We aimed to elucidate the effects of oxidized low-density lipoprotein (OxLDL) on platelet-monocyte interactions and the consequences of these interactions on platelet phagocytosis, chemokine release, monocyte extravasation, and foam cell formation. APPROACH AND RESULTS: Confocal microscopy and flow cytometric analysis revealed that in vitro and in vivo stimulation with OxLDL resulted in rapid formation of platelet-monocyte aggregates, with a preference for CD16+ monocyte subsets. This platelet-monocyte interaction facilitated OxLDL uptake by monocytes, in a process that involved platelet CD36-OxLDL interaction, release of chemokines, such as CXC motif ligand 4, direct platelet-monocyte interaction, and phagocytosis of platelets. Inhibition of cyclooxygenase with acetylsalicylic acid and antagonists of ADP receptors, P2Y1 and P2Y12, partly abrogated OxLDL-induced platelet-monocyte aggregates and platelet-mediated lipid uptake in monocytes. Platelets also enhanced OxLDL-induced monocyte transmigration across an endothelial monolayer via direct interaction with monocytes in a transwell assay. Importantly, in LDLR(-/-) mice, platelet depletion resulted in a significant decrease of peritoneal macrophage recruitment and foam cell formation in a thioglycollate-elicited peritonitis model. In platelet-depleted wild-type mice, transfusion of ex vivo OxLDL-stimulated platelets induced monocyte extravasation to a higher extent when compared with resting platelets. CONCLUSIONS: Our results on OxLDL-mediated platelet-monocyte aggregate formation, which promoted phenotypic changes in monocytes, monocyte extravasation and enhanced foam cell formation in vitro and in vivo, provide a novel mechanism for how platelets potentiate key steps of atherosclerotic plaque development and plaque destabilization.


Asunto(s)
Plaquetas/fisiología , Quimiotaxis/efectos de los fármacos , Células Espumosas/efectos de los fármacos , Lipoproteínas LDL/farmacología , Monocitos/efectos de los fármacos , Animales , Aspirina/farmacología , Aterosclerosis/fisiopatología , Plaquetas/citología , Plaquetas/efectos de los fármacos , Antígeno CD11b/fisiología , Células Cultivadas , Quimiocinas/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Células Espumosas/citología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipoproteínas LDL/toxicidad , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Selectina-P/sangre , Peritonitis/inducido químicamente , Peritonitis/patología , Fagocitosis/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Factor Plaquetario 4/fisiología , Transfusión de Plaquetas , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores de LDL/deficiencia , Receptores de LDL/genética , Migración Transendotelial y Transepitelial/efectos de los fármacos
16.
Arterioscler Thromb Vasc Biol ; 34(4): 801-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24558109

RESUMEN

OBJECTIVE: Human cytomegalovirus (HCMV) is a widespread pathogen that correlates with various clinical complications, including atherosclerosis. HCMV is released into the circulation during primary infection and periodic viral reactivation, allowing virus-platelet interactions. Platelets are important in the onset and development of atherosclerosis, but the consequences of platelet-HCMV interactions are unclear. APPROACH AND RESULTS: We studied the effects of HCMV-platelet interactions in blood from healthy donors using the purified clinical HCMV isolate VR1814. We demonstrated that HCMV bound to a Toll-like receptor (TLR) 2-positive platelet subpopulation, which resulted in signal transduction, degranulation, and release of proinflammatory CD40L and interleukin-1ß and proangiogenic vascular endothelial-derived growth factor. In mice, murine CMV activated wild-type but not TLR2-deficient platelets. However, supernatant from murine CMV-stimulated wild-type platelets also activated TLR2-deficient platelets, indicating that activated platelets generated soluble mediators that triggered further platelet activation, independent of TLR2 expression. Inhibitor studies, using ADP receptor antagonists and apyrase, revealed that ADP release is important to trigger secondary platelet activation in response to HCMV. HCMV-activated platelets rapidly bound to and activated neutrophils, supporting their adhesion and transmigration through endothelial monolayers. In an in vivo model, murine CMV induced systemic upregulation of platelet-leukocyte aggregates and plasma vascular endothelial-derived growth factor in mice and showed a tendency to enhance neutrophil extravasation in a TLR2-dependent fashion. CONCLUSIONS: HCMV is a well-adapted pathogen that does not induce immediate thrombotic events. However, HCMV-platelet interactions lead to proinflammatory and proangiogenic responses, which exacerbate tissue damage and contribute to atherogenesis. Therefore, platelets might contribute to the effects of HCMV in accelerating atherosclerosis.


Asunto(s)
Plaquetas/metabolismo , Plaquetas/virología , Citomegalovirus/patogenicidad , Mediadores de Inflamación/metabolismo , Inflamación/etiología , Neovascularización Patológica , Receptor Toll-Like 2/metabolismo , Adenosina Difosfato/metabolismo , Animales , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/virología , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Ligando de CD40/metabolismo , Degranulación de la Célula , Células Cultivadas , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/virología , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Activación Neutrófila , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/virología , Activación Plaquetaria/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2/efectos de los fármacos , Receptores Purinérgicos P2/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Migración Transendotelial y Transepitelial , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
J Immunol ; 190(2): 678-88, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23233723

RESUMEN

Psoriasis is characterized by a specific microRNA expression profile, distinct from that of healthy skin. MiR-31 is one of the most highly overexpressed microRNAs in psoriasis skin; however, its biological role in the disease has not been studied. In this study, we show that miR-31 is markedly overexpressed in psoriasis keratinocytes. Specific inhibition of miR-31 suppressed NF-κB-driven promoter luciferase activity and the basal and TNF-α-induced production of IL-1ß, CXCL1/growth-related oncogene-α, CXCL5/epithelial-derived neutrophil-activating peptide 78, and CXCL8/IL-8 in human primary keratinocytes. Moreover, interference with endogenous miR-31 decreased the ability of keratinocytes to activate endothelial cells and attract leukocytes. By microarray expression profiling, we identified genes regulated by miR-31 in keratinocytes. Among these genes, we identified serine/threonine kinase 40 (STK40), a negative regulator of NF-κB signaling, as a direct target for miR-31. Silencing of STK40 rescued the suppressive effect of miR-31 inhibition on cytokine/chemokine expression, indicating that miR-31 regulates cytokine/chemokine expression via targeting STK40 in keratinocytes. Finally, we demonstrated that TGF-ß1, a cytokine highly expressed in psoriasis epidermis, upregulated miR-31 expression in keratinocytes in vitro and in vivo. Collectively, our findings suggest that overexpression of miR-31 contributes to skin inflammation in psoriasis lesions by regulating the production of inflammatory mediators and leukocyte chemotaxis to the skin. Our data indicate that inhibition of miR-31 may be a potential therapeutic option in psoriasis.


Asunto(s)
Citocinas/biosíntesis , Expresión Génica , Queratinocitos/metabolismo , MicroARNs/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Psoriasis/genética , Psoriasis/inmunología , Quimiocinas/biosíntesis , Quimiotaxis de Leucocito/inmunología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , FN-kappa B , Proteínas Serina-Treonina Quinasas/genética , Psoriasis/enzimología , Interferencia de ARN , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
18.
J Am Soc Nephrol ; 25(8): 1669-78, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24722450

RESUMEN

Anemia is a feature of CKD and a complication of renal transplantation, often caused by impaired production of erythropoietin. The kidney is a target organ for human cytomegalovirus (hCMV) in such patients, but it is not known whether hCMV effects erythropoietin production. We found that kidneys from patients with CKD were positive for hCMV protein and that blood levels of hCMV IgG inversely correlated with red blood cell count. In mice, systemic murine cytomegalovirus infection decreased serum erythropoietin levels. In human erythropoietin-producing cells, hCMV inhibited hypoxia-induced expression of erythropoietin mRNA and protein. hCMV early gene expression was responsible, as ultraviolet-inactivated virus had no effect and valganciclovir treatment showed that late gene expression was nonessential. Hypoxia-induced gene transcription is controlled by the transcription factors hypoxia-inducible transcription factor (HIF)-1α and HIF2α, which are constitutively produced but stable only under low oxygen conditions. We found that hCMV inhibited constitutive production of HIF2α mRNA. HIF2α is thought to be the master regulator of erythropoietin transcription. Single-cell analysis revealed that nuclear accumulation of HIF2α was inhibited in hCMV-infected cells, and the extent of inhibition correlated with hCMV protein expression. Our findings suggest that renal hCMV infection could induce or exacerbate anemia in patients.


Asunto(s)
Citomegalovirus/fisiología , Eritropoyetina/metabolismo , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/virología , Animales , Anticuerpos Antivirales/sangre , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Técnicas de Cultivo de Célula , Hipoxia de la Célula , Recuento de Eritrocitos , Eritropoyetina/genética , Hemoglobinas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunoglobulina G/metabolismo , Ratones , ARN Mensajero/metabolismo , Insuficiencia Renal Crónica/patología
19.
Carcinogenesis ; 35(1): 145-54, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23978378

RESUMEN

A lack of gap junctional intercellular communication (GJIC) is common in cancer. Many oncogenic viruses have been shown to downregulate the junctional protein connexin 43 (Cx43) and reduce GJIC. Human cytomegalovirus (HCMV) is a ubiquitous, species-specific betaherpesvirus that establishes life-long latency after primary infection. It encodes two viral gene products, immediate early (IE) proteins IE1 and IE2, which are crucial in viral replication and pathogenesis of many diseases. Emerging evidence demonstrates that HCMV DNA and proteins are highly prevalent in glioblastoma multiforme (GBM) and in other tumors, but HCMV's role in tumorigenesis remains obscure. In the present study, we examined the effects of HCMV infection on Cx43 expression and GJIC as well as the viral mechanism mediating the effects in human GBM cells and tissue samples. We found that HCMV downregulated Cx43 protein, resulting in disruption of functional GJIC as assayed by fluorescent dye transfer assay. We show that both HCMV-IE72 and IE86 mediate downregulation of Cx43 by silencing RNA targeting either IE72 or IE86 coupled with ganciclovir. This finding was further validated by transfection with expression vectors encoding IE72 or IE86, and we show that viral-mediated Cx43 depletion involved proteasomal degradation. Importantly, we also observed that the Cx43 protein levels and IE staining correlated inversely in 10 human GBM tissue specimens. Thus, HCMV regulates Cx43 expression and GJIC, which may contribute to gliomagenesis.


Asunto(s)
Neoplasias del Sistema Nervioso Central/virología , Conexina 43/metabolismo , Infecciones por Citomegalovirus/metabolismo , Uniones Comunicantes/metabolismo , Glioblastoma/virología , Proteínas Inmediatas-Precoces/metabolismo , Transactivadores/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Conexina 43/genética , Citomegalovirus/metabolismo , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/virología , Fibroblastos/metabolismo , Fibroblastos/virología , Glioblastoma/patología , Humanos , Proteínas Inmediatas-Precoces/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Transactivadores/genética , Células Tumorales Cultivadas
20.
J Virol ; 87(1): 67-79, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23055571

RESUMEN

Macrophages (MΦ) are first targets during human cytomegalovirus (HCMV) infection and are thought to be crucial for viral persistence and dissemination. However, since MΦ are also a first line of defense and key modulators of the immune response, these cells are at the crossroad between protection and viral pathogenesis. To date, the MΦ-specific contribution to the immune response against HCMV is still poorly understood. In view of the opposite roles of M1 and M2 MΦ during initiation and resolution of the immune response, we characterized the effects of HCMV infection on classically activated M1 MΦ and alternatively activated M2 MΦ. Although HCMV susceptibility was higher in M2 MΦ, HCMV established a productive and persistent infection in both types of MΦ. Upon HCMV encounter, both types of MΦ acquired similar features of classical activation and secreted high levels of proinflammatory cytokines and chemokines. As a functional consequence, conditioned media obtained from HCMV-infected M1 and M2 MΦ potently activated freshly isolated monocytes. Finally, compared to HCMV-infected monocyte-derived dendritic cells, infected M1 and M2 MΦ were more efficient in stimulating proliferation of autologous T cells from HCMV-seropositive donors at early times (24 h) postinfection, while the MΦ immunostimulatory properties were reduced, but not abrogated, at later times (72 h postinfection). In summary, our findings indicate that MΦ preserve proper antigen presentation capacity upon HCMV infection while enhancing inflammation, thus suggesting that MΦ play a role in the maintenance of the large HCMV-specific T-cell repertoire in seropositive individuals.


Asunto(s)
Proliferación Celular , Infecciones por Citomegalovirus/inmunología , Macrófagos/inmunología , Macrófagos/virología , Linfocitos T/inmunología , Células Cultivadas , Citocinas/metabolismo , Humanos , Inflamación , Linfocitos T/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA