Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(27): 8427-32, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26080445

RESUMEN

The use of receptor-ligand interactions to direct toxins to kill diseased cells selectively has shown considerable promise for treatment of a number of cancers and, more recently, autoimmune disease. Here we move the fusion toxin protein (FTP) technology beyond cancer/autoimmune therapeutics to target the human viral pathogen, human cytomegalovirus (HCMV), on the basis of its expression of the 7TM G protein-coupled chemokine receptor US28. The virus origin of US28 provides an exceptional chemokine-binding profile with high selectivity and improved binding for the CX3C chemokine, CX3CL1. Moreover, US28 is constitutively internalizing by nature, providing highly effective FTP delivery. We designed a synthetic CX3CL1 variant engineered to have ultra-high affinity for US28 and greater specificity for US28 than the natural sole receptor for CX3CL1, CX3CR1, and we fused the synthetic variant with the cytotoxic domain of Pseudomonas Exotoxin A. This novel strategy of a rationally designed FTP provided unparalleled anti-HCMV efficacy and potency in vitro and in vivo.


Asunto(s)
Proteínas Bacterianas/metabolismo , Quimiocina CX3CL1/metabolismo , Infecciones por Citomegalovirus/prevención & control , Receptores de Quimiocina/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacología , Proteínas Virales/antagonistas & inhibidores , Proteínas Bacterianas/genética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Quimiocina CX3CL1/genética , Citomegalovirus/genética , Citomegalovirus/metabolismo , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/virología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/virología , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Pulmón/citología , Unión Proteica/efectos de los fármacos , Receptores de Quimiocina/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Proteínas Virales/metabolismo
2.
J Gen Virol ; 94(Pt 5): 1111-1120, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23303826

RESUMEN

Human cytomegalovirus (HCMV)-encoded G protein-coupled-receptor US28 is believed to participate in virus dissemination through modulation of cell migration and immune evasion. US28 binds different CC chemokines and the CX3C chemokine CX3CL1. Membrane-anchored CX3CL1 is expressed by immune-activated endothelial cells, causing redirection of CX3CR1-expressing leukocytes in the blood to sites of infection. Here, we used stable transfected cell lines to examine how US28 expression affects cell migration on immobilized full-length CX3CL1, to model how HCMV-infected leukocytes interact with inflamed endothelium. We observed that US28-expressing cells migrated more than CX3CR1-expressing cells when adhering to immobilized CX3CL1. US28-induced migration was G protein-signalling dependent and was blocked by the phospholipase Cß inhibitor U73122 and the intracellular calcium chelator BAPTA-AM. In addition, migration was inhibited in a dose-dependent manner by competition from CCL2 and CCL5, whereas CCL3 had little effect. Instead of migrating, CX3CR1-expressing cells performed 'dancing-on-the-spot' movements, demonstrating that anchored CX3CL1 acts as a strong tether for these cells. At low receptor expression levels, however, no significant difference in migration potential was observed when comparing the migration of CX3CR1- and US28-expressing cells. Thus, these data showed that, in contrast to CX3CR1, which promotes efficient cell capture upon binding to anchored CX3CL1, US28 acts to increase the migration of cells upon binding to the same ligand. Overall, this indicates that infected cells probably move more than uninfected cells in inflamed tissues with high CX3CL1 expression, with soluble chemokines affecting the final migration.


Asunto(s)
Movimiento Celular , Quimiocina CX3CL1/metabolismo , Infecciones por Citomegalovirus/virología , Citomegalovirus/metabolismo , Receptores de Quimiocina/metabolismo , Proteínas Virales/metabolismo , Receptor 1 de Quimiocinas CX3C , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quelantes/farmacología , Quimiocina CX3CL1/genética , Quimiocinas CC/metabolismo , Relación Dosis-Respuesta a Droga , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Células Endoteliales , Estrenos/farmacología , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Inhibidores de Fosfodiesterasa/farmacología , Fosfolipasa C beta/antagonistas & inhibidores , Pirrolidinonas/farmacología , Receptores de Quimiocina/genética , Transducción de Señal , Imagen de Lapso de Tiempo , Proteínas Virales/genética
3.
J Virol ; 85(12): 6091-5, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21490099

RESUMEN

The human cytomegalovirus (CMV) proteins US28 and UL33 are homologous to chemokine receptors (CKRs). Knockout of the mouse CMV M33 protein (UL33 homologue) results in substantial attenuation of salivary gland infection/replication and reduced efficiency of reactivation from tissue explants. M33-mediated G protein-coupled signaling is critical for the salivary gland phenotype. In this report, we demonstrate that US28 and (to a lesser degree) UL33 restore reactivation from tissue explants and partially restore replication in salivary glands (compared to a signaling-deficient M33 mutant). These studies provide a novel small animal model for evaluation of therapies targeting the human CMV CKRs.


Asunto(s)
Citomegalovirus/fisiología , Modelos Animales de Enfermedad , Muromegalovirus/fisiología , Receptores de Quimiocina/metabolismo , Proteínas Virales/metabolismo , Animales , Citomegalovirus/genética , Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Femenino , Infecciones por Herpesviridae/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Muromegalovirus/genética , Muromegalovirus/metabolismo , Especificidad de Órganos , Receptores de Quimiocina/genética , Glándulas Salivales/metabolismo , Glándulas Salivales/virología , Proteínas Virales/genética , Activación Viral , Latencia del Virus , Replicación Viral
4.
Ugeskr Laeger ; 184(24)2022 06 13.
Artículo en Danés | MEDLINE | ID: mdl-35703076

RESUMEN

Viroporins are ion channels found in many viruses, where they contribute to virus life cycle and thereby pathogenesis. Viroporin targeting is a known, yet largely unexplored, therapeutic strategy so far only used in Influenza A with the drugs amantadine and rimantadine. In this review, we seek to utilize the inhibition by amantadine of the viroporin Protein E in SARS-CoV-2 in an attempt to treat COVID-19 in its early stages. We are executing a double-blinded placebo-controlled trial based on promising in vivo and in vitro work as a stepping-stone for establishing a therapeutic antiviral regime: blocking of viroporins.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Amantadina , Antivirales/farmacología , Antivirales/uso terapéutico , Humanos , SARS-CoV-2 , Proteínas Viroporinas
5.
J Heart Lung Transplant ; 41(3): 287-297, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34802874

RESUMEN

BACKGROUND: Transmission of latent human cytomegalovirus (HCMV) via organ transplantation with post-transplant viral reactivation is extremely prevalent and results in substantial adverse impact on outcomes. Therapies targeting the latent reservoir within the allograft to mitigate viral transmission would represent a major advance. Here, we delivered an immunotoxin (F49A-FTP) that targets and kills latent HCMV aiming at reducing the HCMV reservoir from donor lungs using ex-vivo lung perfusion (EVLP). METHODS: HCMV seropositive human lungs were placed on EVLP alone or EVLP + 1mg/L of F49A-FTP for 6 hours (n = 6, each). CD14+ monocytes isolated from biopsies pre and post EVLP underwent HCMV reactivation assay designed to evaluate viral reactivation capacity. Off-target effects of F49A-FTP were studied evaluating cell death markers of CD34+ and CD14+ cells using flow cytometry. Lung function on EVLP and inflammatory cytokine production were evaluated as safety endpoints. RESULTS: We demonstrate that lungs treated ex-vivo with F49A-FTP had a significant reduction in HCMV reactivation compared to controls, suggesting successful targeting of latent virus (76% median reduction in F49A-FTP vs 15% increase in controls, p = 0.0087). Furthermore, there was comparable cell death rates of the targeted cells between both groups, suggesting no off-target effects. Ex-vivo lung function was stable over 6 hours and no differences in key inflammatory cytokines were observed demonstrating safety of this novel treatment. CONCLUSIONS: Ex-vivo F49A-FTP treatment of human lungs targets and kills latent HCMV, markedly attenuating HCMV reactivation. This approach demonstrates the first experiments targeting latent HCMV in a donor organ with promising results towards clinical translation.


Asunto(s)
Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Inmunotoxinas/farmacología , Inmunotoxinas/uso terapéutico , Trasplante de Pulmón , Selección de Paciente , Quimiocina CX3CL1 , Exotoxinas , Humanos , Técnicas In Vitro
6.
mBio ; 10(1)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30647152

RESUMEN

The Epstein-Barr virus (EBV) BILF1 gene encodes a constitutively active G protein-coupled receptor (GPCR) that downregulates major histocompatibility complex (MHC) class I and induces signaling-dependent tumorigenesis. Different BILF1 homologs display highly conserved extracellular loops (ECLs) including the conserved cysteine residues involved in disulfide bridges present in class A GPCRs (GPCR bridge between transmembrane helix 3 [TM-3] and ECL-2) and in chemokine receptors (CKR bridge between the N terminus and ECL-3). In order to investigate the roles of the conserved residues in the receptor functions, 25 mutations were created in the extracellular domains. Luciferase reporter assays and flow cytometry were used to investigate the G protein signaling and MHC class I downregulation in HEK293 cells. We find that the cysteine residues involved in the GPCR bridge are important for both signaling and MHC class I downregulation, whereas the cysteine residues in the N terminus and ECL-3 are dispensable for signaling but important for MHC class I downregulation. Multiple conserved residues in the extracellular regions are important for the receptor-induced MHC class I downregulation, but not for signaling, indicating distinct structural requirements for these two functions. In an engineered receptor containing a binding site for Zn+2 ions in a complex with an aromatic chelator (phenanthroline or bipyridine), a ligand-driven inhibition of both the receptor signaling and MHC class I downregulation was observed. Taken together, this suggests that distinct regions in EBV-BILF1 can be pharmacologically targeted to inhibit the signaling-mediated tumorigenesis and interfere with the MHC class I downregulation.IMPORTANCE G protein-coupled receptors constitute the largest family of membrane proteins. As targets of >30% of the FDA-approved drugs, they are valuable for drug discovery. The receptor is composed of seven membrane-spanning helices and intracellular and extracellular domains. BILF1 is a receptor encoded by Epstein-Barr virus (EBV), which evades the host immune system by various strategies. BILF1 facilitates the virus immune evasion by downregulating MHC class I and is capable of inducing signaling-mediated tumorigenesis. BILF1 homologs from primate viruses show highly conserved extracellular domains. Here, we show that conserved residues in the extracellular domains of EBV-BILF1 are important for downregulating MHC class I and that the receptor signaling and immune evasion can be inhibited by drug-like small molecules. This suggests that BILF1 could be a target to inhibit the signaling-mediated tumorigenesis and interfere with the MHC class I downregulation, thereby facilitating virus recognition by the immune system.


Asunto(s)
Regulación hacia Abajo , Herpesvirus Humano 4/fisiología , Antígenos de Histocompatibilidad Clase I/biosíntesis , Interacciones Huésped-Patógeno , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Proteínas Virales/metabolismo , Análisis Mutacional de ADN , Citometría de Flujo , Genes Reporteros , Células HEK293 , Herpesvirus Humano 4/genética , Humanos , Luciferasas/análisis , Luciferasas/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Receptores Acoplados a Proteínas G/genética , Proteínas Virales/genética
7.
J Immunol Res ; 2017: 4069260, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28251165

RESUMEN

Immunotoxins as antiviral therapeutics are largely unexplored but have promising prospective due to their high selectivity potential and their unparalleled efficiency. One recent example targeted the virus-encoded G protein-coupled receptor US28 as a strategy for specific and efficient treatment of human cytomegalovirus (HCMV) infections. US28 is expressed on virus-infected cells and scavenge chemokines by rapid internalization. The chemokine-based fusion-toxin protein (FTP) consisted of a variant (F49A) of CX3CL1 specifically targeting US28 linked to the catalytic domain of Pseudomonas exotoxin A (PE). Here, we systematically seek to improve F49A-FTP by modifications in its three structural domains; we generated variants with (1) altered chemokine sequence (K14A, F49L, and F49E), (2) shortened and elongated linker region, and (3) modified toxin domain. Only F49L-FTP displayed higher selectivity in its binding to US28 versus CX3CR1, the endogenous receptor for CX3CL1, but this was not matched by a more selective killing of US28-expressing cells. A longer linker and different toxin variants decreased US28 affinity and selective killing. Thereby, F49A-FTP represents the best candidate for HCMV treatment. Many viruses encode internalizing receptors suggesting that not only HCMV but also, for instance, Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus may be targeted by FTPs.


Asunto(s)
Antivirales/inmunología , Antivirales/uso terapéutico , Citomegalovirus/inmunología , Inmunotoxinas/uso terapéutico , Antivirales/efectos adversos , Proteínas Bacterianas/inmunología , Células Cultivadas , Quimiocina CX3CL1/metabolismo , Quimiocina CX3CL1/uso terapéutico , Fibroblastos , Herpesvirus Humano 4 , Humanos , Inmunotoxinas/efectos adversos , Estudios Prospectivos , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Transducción de Señal , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
8.
Curr Drug Targets ; 7(1): 103-18, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16454703

RESUMEN

Viral infections depend on an intimate relationship between the infectious agent and the host cells. Viruses need the host cells for replication, while the innate- and adaptive- immunesystem of the host is fighting to kill the infected cell in order to clear out the pathogen and survive the infection. However, since both virus and host exist, the organisms struggle must reach an ecological equilibrium. Among the best-studied interactions between viruses and the host immune system are those between herpesviruses and their hosts. Herpesviruses are known to devote a significant part of their large genomes on immuno-modulatory genes, some encoding chemokines or chemokine receptors. These genes, which may be dispensable for viral replication in vitro, are highly important for viral growth in vivo, for viral dissemination and disease progression. Indeed, all beta and gamma-herpesviruses have acquired homologs of both chemokines and chemokine receptors belonging to the 7 transmembrane (7TM) spanning, G protein-coupled receptor family. 7TM receptors are very efficient drug targets and are currently the most popular class of investigational drug targets. A notable trait for the virus encoded chemokine receptors seems to be their constitutive activity. The biological function of the constitutive activity is still unclear, but it has become clear that the receptors are involved in important parts of the viral lifecycle in vivo, and that the receptor signaling is involved in gamma-herpesvirus mediated cell transformation. Therefore, blocking the signaling of these receptors will provide an efficient and highly specific way to inhibit viral replication in vivo and disease progression in the hosts.


Asunto(s)
Quimiocinas/fisiología , Infecciones por Herpesviridae/tratamiento farmacológico , Infecciones por Herpesviridae/fisiopatología , Animales , Quimiocinas/biosíntesis , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/fisiopatología , Herpesviridae/fisiología , Infecciones por Herpesviridae/virología , Humanos , Receptores de Quimiocina/biosíntesis , Receptores de Quimiocina/fisiología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/fisiología
9.
J Leukoc Biol ; 99(6): 911-25, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26729815

RESUMEN

There is a constant need for new therapeutic interventions in a wide range of infectious diseases. Over the past few years, the immunotoxins have entered the stage as promising antiviral treatments. Immunotoxins have been extensively explored in cancer treatment and have achieved FDA approval in several cases. Indeed, the design of new anticancer immunotoxins is a rapidly developing field. However, at present, several immunotoxins have been developed targeting a variety of different viruses with high specificity and efficacy. Rather than blocking a viral or cellular pathway needed for virus replication and dissemination, immunotoxins exert their effect by killing and eradicating the pool of infected cells. By targeting a virus-encoded target molecule, it is possible to obtain superior selectivity and drastically limit the side effects, which is an immunotoxin-related challenge that has hindered the success of immunotoxins in cancer treatment. Therefore, it seems beneficial to use immunotoxins for the treatment of virus infections. One recent example showed that targeting of virus-encoded 7 transmembrane (7TM) receptors by immunotoxins could be a future strategy for designing ultraspecific antiviral treatment, ensuring efficient internalization and hence efficient eradication of the pool of infected cells, both in vitro and in vivo. In this review, we provide an overview of the mechanisms of action of immunotoxins and highlight the advantages of immunotoxins as future anti-viral therapies.


Asunto(s)
Antivirales/metabolismo , Inmunotoxinas/metabolismo , Animales , Quimiocinas/metabolismo , Citomegalovirus/fisiología , Humanos , Modelos Biológicos , Receptores de Quimiocina/metabolismo , Proteínas Virales/metabolismo
10.
Circulation ; 109(4): 500-5, 2004 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-14732750

RESUMEN

BACKGROUND: We hypothesized that cytomegalovirus (CMV) may contribute to the vasculopathy observed in cardiac allograft recipients by impairing the endothelial nitric oxide synthase pathway. We focused on asymmetric dimethylarginine (ADMA, the endogenous inhibitor of nitric oxide synthase) as a potential mediator of the adverse vascular effect of CMV. METHODS AND RESULTS: Heart transplant recipients manifested elevated plasma ADMA levels compared with healthy control subjects. Transplant patients with CMV DNA-positive leukocytes had higher plasma ADMA concentrations and more extensive transplant arteriopathy (TA). Human microvascular endothelial cells infected with the CMV isolates elaborated more ADMA. The increase in ADMA was temporally associated with a reduction in the activity of dimethylarginine dimethylaminohydrolase (DDAH, the enzyme that metabolizes ADMA). Infected cultures showed high levels of oxidative stress with enhanced endothelial production of superoxide anion. CONCLUSIONS: CMV infection in human heart transplant recipients is associated with higher ADMA elevation and more severe TA. CMV infection in endothelial cells increases oxidative stress, impairs DDAH activity, and increases ADMA elaboration. CMV infection may contribute to endothelial dysfunction and TA by dysregulation of the endothelial nitric oxide synthase pathway.


Asunto(s)
Arginina/análogos & derivados , Arginina/sangre , Enfermedad de la Arteria Coronaria/virología , Infecciones por Citomegalovirus/complicaciones , Trasplante de Corazón/efectos adversos , Óxido Nítrico Sintasa/metabolismo , Amidohidrolasas/metabolismo , Arginina/metabolismo , Arginina/fisiología , Enfermedades Cardiovasculares/epidemiología , Células Cultivadas , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/etiología , GMP Cíclico/biosíntesis , Infecciones por Citomegalovirus/enzimología , Endotelio Vascular/metabolismo , Endotelio Vascular/virología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nitritos/metabolismo , Estrés Oxidativo , Factores de Riesgo , Transducción de Señal
11.
Biomaterials ; 30(29): 5305-11, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19577290

RESUMEN

Microcontact printing (mCP) is employed to generate discontinuous microscale gradients of active fractalkine, a chemokine expressed by endothelial cells near sites of inflammation where it is believed to form concentration gradients descending away from the inflamed area. In vivo, fractalkine is a transmembrane molecule extending its chemokine domain into the vascular lumen. Substrate bound in vitro gradients may thus closely resemble in vivo conditions. Direct mCP of sensitive proteins like fractalkine may cause partial protein denaturation and will not ensure correct orientation of the biologically active part of the molecules. Here, indirect mCP of a capture antibody recognizing a molecular tag on the target protein is successfully used to pattern tagged fractalkine in microscale gradient patterns. Fractalkine functions as an adhesion molecule for leukocytes. Cells expressing the fractalkine receptor are found to attach to the gradient structure at a density correlated with the fractional area covered by fractalkine. This indicates that the patterned fractalkine maintains its biological function. The method can be applied to in vitro studies of cell responses to the wide range of naturally surface-bound chemokines (haptotactic gradients). The use of a capture antibody facilitates control of the orientation of tagged molecules, thereby ensuring a high degree of bio-functionality through correct presentation and reduced protein denaturation.


Asunto(s)
Quimiocina CX3CL1/química , Quimiocina CX3CL1/farmacología , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Riñón/citología , Riñón/fisiología , Adsorción , Adhesión Celular/efectos de los fármacos , Línea Celular , Humanos , Riñón/efectos de los fármacos , Ensayo de Materiales
12.
J Biol Chem ; 281(19): 13199-13208, 2006 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-16540462

RESUMEN

Epstein-Barr virus (EBV)-induced receptor 2 (EBI2) is an orphan seven-transmembrane (7TM) receptor originally identified as the most up-regulated gene (>200-fold) in EBV-infected cells. Here we show that EBI2 signals with constitutive activity through Galpha(i) as determined by a receptor-mediated inhibition of forskolin-induced cAMP production and an induction of the serum response element-driven transcriptional activity in a pertussis toxin-sensitive manner. Galpha(s) and Galpha(q) were not activated constitutively as determined by the lack of cAMP production, the lack of inositol phosphate turnover, and the lack of activities of the transcription factors: cAMP response element-binding protein and nuclear factor-kappaB. Immunohistochemistry and confocal microscopy of FLAG- and green fluorescent protein-tagged EBI2 revealed cell-surface expression. A putative N-terminal truncated version of EBI2, delta4-EBI2, showed similar expression and signaling through Galpha(i) as full-length EBI2. By using a 32P-labeled EBI2 probe we found a very high expression in lymphoid tissue (spleen and lymph node) and peripheral blood mononuclear cells and a high expression in lung tissue. Real-time PCR of EBV-infected cells showed high expression of EBI2 during latent and lytic infection, in contrast to the EBV-encoded 7TM receptor BILF1, which was induced during lytic infection. EBI2 clustered with the orphan GPR18 by alignment analysis as well as by close proximity in the chromosomal region 13q32.3. Based on the constitutive signaling and cellular expression pattern of EBI2, it is suggested that it may function in conjunction with BILF1 in the reprogramming of the cell during EBV infection.


Asunto(s)
Receptores de Superficie Celular/metabolismo , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Humanos , Células Asesinas Naturales/metabolismo , Linfocitos/metabolismo , Monocitos/metabolismo , Isoformas de Proteínas , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
13.
Mol Pharmacol ; 68(1): 11-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15788740

RESUMEN

The highly conserved Arg in the so-called DRY motif (Asp-Arg-Tyr) at the intracellular end of transmembrane helix 3 is in general considered as an essential residue for G protein coupling in rhodopsin-like seven transmembrane (7TM) receptors. In the open reading frame 74 (ORF74) receptor encoded by equine herpesvirus 2 (EHV2), the DRY motif is substituted with a DTW motif. Nevertheless, this receptor signaled with high constitutive activity through Gi as determined by a receptor-mediated inhibition of forskolin-induced cAMP-production and by an induction of the serum response element-driven transcriptional activity through a pertussis toxin-sensitive manner. Gs and Gq were not activated constitutively as determined by the lack of inositol phosphate turnover and activities of the three transcription factors: cAMP response element-binding protein (CREB), nuclear factor-kappaB, and nuclear factor of activated T cells. Coexpression of the ORF74-EHV2 receptor with the promiscuous G protein Gqi4myr supported the constitutive Gi activation as determined by inositol phosphate turnover and CREB activation. The constitutive activity was inhibited by nonpeptide inverse agonists with micromolar potencies, and the chemokine CXCL6 acted as a high-affinity agonist. It is noteworthy that reconstitution of the DRY motif resulted in a 4- to 5-fold decrease of the constitutive activity. Both the wild type and the receptor with the reconstituted DRY motif were expressed at the cell surface as indicated by immunohistochemistry and enzyme-linked immunosorbent assay analysis. It is concluded that the Arg of the DRY motif in transmembrane helix 3 is not essential for G protein coupling based on the constitutive as well as the ligand-mediated activity observed for ORF74-EHV2.


Asunto(s)
Arginina/genética , Ácido Aspártico/genética , Membrana Celular/genética , Receptores de Quimiocina/genética , Receptores Acoplados a Proteínas G/genética , Rhadinovirus/genética , Tirosina/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Arginina/química , Arginina/metabolismo , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Secuencia Conservada , Relación Dosis-Respuesta a Droga , Humanos , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Receptores de Quimiocina/química , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Rhadinovirus/química , Tirosina/química , Tirosina/metabolismo
14.
J Virol ; 79(1): 536-46, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15596846

RESUMEN

Both beta- and gammaherpesviruses encode G protein-coupled receptors (GPCRs) with unique pharmacological phenotypes and important biological functions. An example is ORF74, the gamma2-herpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV)-encoded GPCR, which is highly constitutively active and considered the key oncogene in Kaposi's sarcoma pathogenesis. In contrast, the current annotation of the Epstein-Barr virus (EBV) genome does not reveal any GPCR homolog encoded by this human oncogenic gamma1-herpesvirus. However, by employing bioinformatics, we recognized that the previously established EBV open reading frame BILF1 indeed encodes a GPCR. Additionally, BILF1 is a member of a new family of related GPCRs exclusively encoded by gamma1-herpesviruses. Expression of hemagglutinin-tagged BILF1 in the HEK293 epithelial cell line revealed that BILF1 is expressed as an approximately 50-kDa glycosylated protein. Immunocytochemistry and confocal microscopy revealed that BILF1 localizes predominantly to the plasma membrane, similar to the localization of KSHV ORF74. Using chimeric G proteins, we found that human and rhesus EBV-encoded BILF1 are highly potent constitutively active receptors, activating Galphai. Furthermore, BILF1 is able to inhibit forskolin-triggered CREB activation via stimulation of endogenous G proteins in a pertussis toxin-sensitive manner, verifying that BILF1 signals constitutively through Galphai. We suggest that EBV may use BILF1 to regulate Galphai-activated pathways during viral lytic replication, thereby affecting disease progression.


Asunto(s)
Herpesvirus Humano 4/patogenicidad , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular , Membrana Celular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Glicosilación , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Inmunohistoquímica , Microscopía Confocal , Datos de Secuencia Molecular , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Proteínas Virales/química , Proteínas Virales/genética
15.
J Virol ; 79(12): 7570-96, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15919911

RESUMEN

Prediction of protein-coding regions and other features of primary DNA sequence have greatly contributed to experimental biology. Significant challenges remain in genome annotation methods, including the identification of small or overlapping genes and the assessment of mRNA splicing or unconventional translation signals in expression. We have employed a combined analysis of compositional biases and conservation together with frame-specific G+C representation to reevaluate and annotate the genome sequences of mouse and rat cytomegaloviruses. Our analysis predicts that there are at least 34 protein-coding regions in these genomes that were not apparent in earlier annotation efforts. These include 17 single-exon genes, three new exons of previously identified genes, a newly identified four-exon gene for a lectin-like protein (in rat cytomegalovirus), and 10 probable frameshift extensions of previously annotated genes. This expanded set of candidate genes provides an additional basis for investigation in cytomegalovirus biology and pathogenesis.


Asunto(s)
Biología Computacional/métodos , Citomegalovirus/genética , Genoma Viral , Muromegalovirus/genética , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Composición de Base , Secuencia Conservada , Bases de Datos Genéticas , Ratones , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Ratas , Análisis de Secuencia de ADN , Proteínas Virales/química
16.
J Virol ; 76(16): 8161-8, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12134021

RESUMEN

Cellular infection by cytomegalovirus (CMV) is associated with very early G-protein-mediated signal transduction and reprogramming of gene expression. Here we investigated the involvement of human CMV (HCMV)-encoded US27, US28, and UL33 receptors as well as murine CMV-encoded M33 transmembrane (7TM) receptors in host cell signaling mechanisms. HCMV-encoded US27 did not show any constitutive activity in any of the studied signaling pathways; in contrast, US28 and M33 displayed ligand-independent, constitutive signaling through the G protein q (Gq)/phospholipase C pathway. In addition, M33 and US28 also activated the transcription factor NF-kappaB as well as the cyclic AMP response element binding protein (CREB) in a ligand-independent, constitutive manner. The use of specific inhibitors indicated that the p38 mitogen-activated protein (MAP) kinase but not the extracellular signal-regulated kinase 1/2-MAP kinase pathway is involved in M33- and US28-mediated CREB activation but not NF-kappaB activation. Interestingly, UL33-the HCMV-encoded structural homologue of M33-was only marginally constitutively active in the Gq/phospholipase C turnover and CREB activation assays and did not show any constitutive activity in the NF-kappaB pathway, where M33 and US28 were highly active. Hence, CMVs appear to have conserved mechanisms for regulating host gene transcription, i.e., constitutive activation of certain kinases and transcription factors through the constitutive activities of 7TM proteins. These data, together with the previous identification of the incorporation of such proteins in the viral envelope, suggest that these proteins could be involved in the very early reprogramming of the host cell during viral infection.


Asunto(s)
Citomegalovirus/metabolismo , Receptores de Quimiocina/metabolismo , Proteínas Virales/metabolismo , Animales , Células COS , Quimiocinas/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citomegalovirus/genética , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositoles/metabolismo , Receptores de Quimiocina/genética , Transducción de Señal , Especificidad de la Especie , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/virología , Proteínas Virales/genética , Proteínas Quinasas p38 Activadas por Mitógenos
17.
Traffic ; 3(3): 218-32, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11886592

RESUMEN

The human cytomegalovirus genome encodes four putative seven transmembrane domain chemokine receptor-like proteins. Although important in viral pathogenesis, little is known about the properties or functions of these proteins. We previously reported that US28 is located in endocytic vesicles and undergoes constitutive endocytosis and recycling. Here we studied the cellular distributions and trafficking of two other human cytomegalovirus chemokine receptor-like proteins, UL33 and US27, in transfected and human cytomegalovirus-infected cells. Immunofluorescence staining indicated that UL33 and US27 are located at the cell surface, although the majority of both proteins was seen in intracellular organelles located in the perinuclear region of the cell. The intracellular pools of UL33 and US27 showed overlap with markers for endocytic organelles. Antibody-feeding experiments indicated that cell surface US27 undergoes endocytosis. By immunogold labeling of cryosections and electron microscopy, UL33 was seen to localize to multivesicular bodies (MVBs or multivesicular endosomes). Electron microscopy analysis of human cytomegalovirus-infected cells showed that most virus particles wrapped individually into short membrane cisternae, although virus particles were also occasionally seen within and budding into MVBs. Electron microscopy immunolocalization of viral UL33 and US27 on ultrathin cryosections of human cytomegalovirus-infected cells showed gold particles over the membranes into which virions were wrapping, in small membrane tubules and vesicles and in MVBs. Labeling of the human cytomegalovirus glycoproteins gB and gH indicated that these proteins were also present in the same membrane structures. This first electron microscopy analysis of human cytomegalovirus assembly using immunolabeling suggests that the localization of UL33, US27 and US28 to endosomes may allow these proteins to be incorporated into the viral membrane during the final stages of human cytomegalovirus assembly.


Asunto(s)
Membrana Celular/metabolismo , Citomegalovirus/metabolismo , Receptores de Quimiocina/biosíntesis , Proteínas Virales/biosíntesis , Proteínas Virales/química , Animales , Línea Celular , Células Cultivadas , ADN Complementario/metabolismo , Endocitosis , Fibroblastos/metabolismo , Fibroblastos/virología , Glicoproteínas/metabolismo , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Inmunohistoquímica , Proteínas Luminiscentes/metabolismo , Masculino , Microscopía Electrónica , Microscopía Fluorescente , Modelos Genéticos , Conejos , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA