Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 7(11): e1002332, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22072961

ABSTRACT

Interferon-induced BST2/Tetherin prevents budding of vpu-deficient HIV-1 by tethering mature viral particles to the plasma membrane. BST2 also inhibits release of other enveloped viruses including Ebola virus and Kaposi's sarcoma associated herpesvirus (KSHV), indicating that BST2 is a broadly acting antiviral host protein. Unexpectedly however, recovery of human cytomegalovirus (HCMV) from supernatants of BST2-expressing human fibroblasts was increased rather than decreased. Furthermore, BST2 seemed to enhance viral entry into cells since more virion proteins were released into BST2-expressing cells and subsequent viral gene expression was elevated. A significant increase in viral entry was also observed upon induction of endogenous BST2 during differentiation of the pro-monocytic cell line THP-1. Moreover, treatment of primary human monocytes with siRNA to BST2 reduced HCMV infection, suggesting that BST2 facilitates entry of HCMV into cells expressing high levels of BST2 either constitutively or in response to exogenous stimuli. Since BST2 is present in HCMV particles we propose that HCMV entry is enhanced via a reverse-tethering mechanism with BST2 in the viral envelope interacting with BST2 in the target cell membrane. Our data suggest that HCMV not only counteracts the well-established function of BST2 as inhibitor of viral egress but also employs this anti-viral protein to gain entry into BST2-expressing hematopoietic cells, a process that might play a role in hematogenous dissemination of HCMV.


Subject(s)
Antigens, CD/metabolism , Cytomegalovirus/physiology , Virus Internalization , Virus Release , Antigens, CD/genetics , Cell Line , Ebolavirus/physiology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , HIV-1/physiology , Herpesvirus 8, Human/physiology , Humans , Monocytes/virology , RNA Interference , RNA, Small Interfering
2.
Cell Host Microbe ; 8(3): 284-91, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20833379

ABSTRACT

Human cytomegalovirus (HCMV) is a significant cause of morbidity and mortality in organ transplant recipients. The use of granulocyte-colony stimulating factor (G-CSF)-mobilized stem cells from HCMV seropositive donors is suggested to double the risk of late-onset HCMV disease and chronic graft-versus-host disease in recipients when compared to conventional bone marrow transplantation with HCMV seropositive donors, although the etiology of the increased risk is unknown. To understand mechanisms of HCMV transmission in patients receiving G-CSF-mobilized blood products, we generated a NOD-scid IL2Rγ(c)(null)-humanized mouse model in which HCMV establishes latent infection in human hematopoietic cells. In this model, G-CSF induces the reactivation of latent HCMV in monocytes/macrophages that have migrated into organ tissues. In addition to establishing a humanized mouse model for systemic and latent HCMV infection, these results suggest that the use of G-CSF mobilized blood products from seropositive donors pose an elevated risk for HCMV transmission to recipients.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Macrophages/virology , Virus Activation , Virus Latency , Animals , Antigens, CD34/analysis , Bone Marrow Cells/immunology , Bone Marrow Cells/virology , Cytokines/blood , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus/isolation & purification , Disease Models, Animal , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/virology , Humans , Mice , Mice, SCID , Monocytes/virology
3.
J Immunol ; 184(6): 3213-22, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20173022

ABSTRACT

Monocytes are a primary target for human CMV (HCMV) infection and are a key cell type responsible for hematogenous dissemination of the virus. Biologically, these cells have a short lifespan of 1-3 d in the circulation, yet infected cells remain viable for weeks despite the lack of viral antiapoptotic gene expression during this period. To understand the mechanism by which HCMV inhibits the initial phase of monocyte apoptosis, we focused on the viral modulation of early prosurvival cell signaling events after infection. We demonstrate in this study that the viral upregulation of the PI3K pathway promotes an early block in apoptosis after infection. Temporal transcriptome and protein analyses revealed Mcl-1, a member of the Bcl-2 family, was transiently induced in a PI3K-dependent manner during the early stages of HCMV infection. In accord with the survival studies, virally induced levels of Mcl-1 expression dissipated to mock levels by 72 h postinfection. Through the use of Mcl-1-specific small interfering RNA, we confirmed the functional role that Mcl-1 plays as a key early regulator of apoptosis in monocytes. Lastly, we showed that HCMV engagement and activation of the epidermal growth factor receptor during viral binding triggered the upregulation of Mcl-1. Overall, our data indicates that activation of the epidermal growth factor receptor/PI3K signaling pathway, via the PI3K-dependent upregulation of Mcl-1, is required to circumvent apoptosis in naturally short-lived monocytes during the early stages of HCMV infection, thus ensuring the early steps in the viral persistence strategy.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Cell Survival/immunology , Cytomegalovirus/immunology , ErbB Receptors/physiology , Macrophages/immunology , Monocytes/immunology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/physiology , Cell Death/immunology , Cells, Cultured , Cytomegalovirus/pathogenicity , Enzyme Induction/immunology , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/physiology , Immunophenotyping , Macrophages/cytology , Macrophages/virology , Monocytes/cytology , Monocytes/enzymology , Monocytes/virology , Myeloid Cell Leukemia Sequence 1 Protein , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology , Up-Regulation/immunology , Virulence/immunology
4.
Virus Res ; 144(1-2): 329-33, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19427341

ABSTRACT

Human cytomegalovirus infection of monocytes stimulates a unique monocyte differentiation reprogramming resulting in polarization towards an M1 pro-inflammatory macrophage that simultaneously exhibits characteristics of an M2 anti-inflammatory macrophage. Our laboratory has previously shown that HCMV infection stimulates monocyte NF-kappaB and PI(3)K activities and now provides evidence that these cellular factors are essential for the HCMV-induced polarization of infected monocytes/macrophages. We find that the induction of NF-kappaB and PI(3)K activities following HCMV infection was required for the initiation of monocyte-to-macrophage differentiation. HCMV-infected monocytes treated with Bay11-7802 (an inhibitor of NF-kappaB activity) or LY294002 [an inhibitor of PI(3)K activity] prior to infection exhibited a small, round and monocyte-like undifferentiated morphology and the lack of CD68 upregulation (a macrophage differentiation marker). Detailed transcriptome analysis revealed 48%, 7% and 31% of HCMV-induced M1-associated genes were dependent on NF-kappaB, PI(3)K or both activities, respectively; while 100% of HCMV-induced M2-associated genes required both NF-kappaB and PI(3)K activities. Functionally, we demonstrated that NF-kappaB and PI(3)K activities were critical for the production of M1- and M2-associated cytokines/chemokines, in HCMV-induced differentiating monocytes. Supernatant from HCMV-infected monocytes pretreated with Bay11-7802 or LY294002 exhibited an 80% and 67% reduction in cell motility-inducing activity. Overall, these data show that HCMV usurps monocyte NF-kappaB and PI(3)K signal transduction pathways to induce the unique polarization of HCMV-infected monocytes needed for the earliest steps in the viral dissemination and persistence strategy.


Subject(s)
Cytomegalovirus/immunology , Monocytes/immunology , Monocytes/virology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cells, Cultured , Gene Expression Profiling , Humans , Signal Transduction
5.
J Immunol ; 181(1): 698-711, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18566437

ABSTRACT

Monocytes are primary targets for human CMV (HCMV) infection and are proposed to be responsible for hematogenous dissemination of the virus. Monocytes acquire different functional traits during polarization to the classical proinflammatory M1 macrophage or the alternative antiinflammatory M2 macrophage. We hypothesized that HCMV induced a proinflammatory M1 macrophage following infection to promote viral dissemination because, biologically, a proinflammatory state provides the tools to drive infected monocytes from the blood into the tissue. To test this hypothesis of monocyte conversion from a normal quiescent phenotype to an inflammatory phenotype, we used Affymetrix Microarray to acquire a transcriptional profile of infected monocytes at a time point our data emphasized is a key temporal regulatory point following infection. We found that HCMV significantly up-regulated 583 (5.2%) of the total genes and down-regulated 621 (5.5%) of the total genes>or=1.5-fold at 4 h postinfection. Further ontology analysis revealed that genes implicated in classical M1 macrophage activation were stimulated by HCMV infection. We found that 65% of genes strictly associated with M1 polarization were up-regulated, while only 4% of genes solely associated with M2 polarization were up-regulated. Analysis of the monocyte chemokinome at the transcriptional level showed that 44% of M1 and 33% of M2 macrophage chemokines were up-regulated. Proteomic analysis using chemokine Ab arrays confirmed the secretion of these chemotactic proteins from HCMV-infected monocytes. Overall, the results identify that the HCMV-infected monocyte transcriptome displayed a unique M1/M2 polarization signature that was skewed toward the classical M1 activation phenotype.


Subject(s)
Cell Differentiation , Cytomegalovirus , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/metabolism , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Differentiation/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Cytomegalovirus/immunology , Gene Expression Profiling , Humans , Macrophages/immunology , Monocytes/immunology , Phenotype , Phosphatidic Acids , Transcription, Genetic/genetics , Up-Regulation , Uridine/analogs & derivatives
6.
J Virol ; 82(2): 1040-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18003728

ABSTRACT

Human cytomegalovirus induces a proinflammatory monocyte following infection, and we have evidence that NF-kappaB and phosphatidylinositol 3-kinase [PI(3)K] are key mediators in this early activation. To begin to address how these signaling pathways are responsible for the rapid activation of infected monocytes, we examined the role that these pathways played in the transcriptome of infected monocytes. Global transcriptional profiling using cDNA microarrays revealed that a significant number of genes, including inflammatory genes, were regulated in an NF-kappaB- and/or PI(3)K-dependent manner, identifying the NF-kappaB and PI(3)K pathways as key cellular control points in the conversion of monocytes to an activated proinflammatory state following HCMV infection.


Subject(s)
Cytomegalovirus/immunology , Gene Expression Profiling , Monocytes/virology , NF-kappa B/physiology , Phosphatidylinositol 3-Kinases/physiology , Cell Line , Humans , Oligonucleotide Array Sequence Analysis
7.
J Virol ; 81(14): 7683-94, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17507481

ABSTRACT

Infected peripheral blood monocytes are proposed to play a key role in the hematogenous dissemination of human cytomegalovirus (HCMV) to tissues, a critical step in the establishment of HCMV persistence and the development of HCMV-associated diseases. We recently provided evidence for a unique strategy involved in viral dissemination: HCMV infection of primary human monocytes promotes their transendothelial migration and differentiation into proinflammatory macrophages permissive for the replication of the original input virus. To decipher the mechanism of hematogenous spread, we focused on the viral dysregulation of early cellular processes involved in transendothelial migration. Here, we present evidence that both phosphatidylinositol 3-kinase [PI(3)K] and NF-kappaB activities were crucial for the HCMV induction of monocyte motility and firm adhesion to endothelial cells. We found that the beta(1) integrins, the beta(2) integrins, intracellular adhesion molecule 1 (ICAM-1), and ICAM-3 were upregulated following HCMV infection and that they played a key role in the firm adhesion of infected monocytes to the endothelium. The viral regulation of adhesion molecule expression is complex, with PI(3)K and NF-kappaB affecting the expression of each adhesion molecule at different stages of the expression cascade. Our data demonstrate key roles for PI(3)K and NF-kappaB signaling in the HCMV-induced cellular changes in monocytes and identify the biological rationale for the activation of these pathways in infected monocytes, which together suggest a mechanism for how HCMV promotes viral spread to and persistence within host organs.


Subject(s)
Cell Adhesion , Cytomegalovirus/physiology , Monocytes/virology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Humans , Monocytes/cytology
8.
J Virol ; 80(23): 11539-55, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16987970

ABSTRACT

Human cytomegalovirus (HCMV) pathogenesis is dependent on the hematogenous spread of the virus to host tissue. While data suggest that infected monocytes are required for viral dissemination from the blood to the host organs, infected endothelial cells are also thought to contribute to this key step in viral pathogenesis. We show here that HCMV infection of endothelial cells increased the recruitment and transendothelial migration of monocytes. Infection of endothelial cells promoted the increased surface expression of cell adhesion molecules (intercellular cell adhesion molecule 1, vascular cell adhesion molecule 1, E-selectin, and platelet endothelial cell adhesion molecule 1), which were necessary for the recruitment of naïve monocytes to the apical surface of the endothelium and for the migration of these monocytes through the endothelial cell layer. As a mechanism to account for the increased monocyte migration, we showed that HCMV infection of endothelial cells increased the permeability of the endothelium. The cellular changes contributing to the increased permeability and increased naïve monocyte transendothelial migration include the disruption of actin stress fiber formation and the decreased expression of lateral junction proteins (occludin and vascular endothelial cadherin). Finally, we showed that the migrating monocytes were productively infected with the virus, documenting that the virus was transferred to the migrating monocyte during passage through the lateral junctions. Together, our results provide evidence for an active role of the infected endothelium in HCMV dissemination and pathogenesis.


Subject(s)
Cell Movement , Cytomegalovirus Infections/pathology , Cytomegalovirus/physiology , Endothelium, Vascular/virology , Monocytes/virology , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/immunology , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Monocytes/cytology , Monocytes/physiology
9.
J Virol ; 78(9): 4444-53, 2004 May.
Article in English | MEDLINE | ID: mdl-15078925

ABSTRACT

Human cytomegalovirus (HCMV) pathogenesis is characterized by multiple organ system involvement due to viral spread to host organs after a cell-associated viremia. The cell type responsible for HCMV dissemination is unknown. Monocytes are the most likely candidate since they are the predominant cell type infected in the blood. However, monocytes are not productive for viral replication and are abortively infected. The results presented here provide a potential answer to this conundrum. We report that primary HCMV infection of monocytes induces transendothelial migration and monocyte-to-macrophage differentiation and that these HCMV-differentiated macrophages are productive for viral replication. Together, our data suggest a novel mechanism for HCMV pathogenesis; HCMV induces cellular changes in monocytes to promote viral replication and spread to host organs.


Subject(s)
Cell Movement , Cytomegalovirus Infections/microbiology , Cytomegalovirus/physiology , Cytomegalovirus/pathogenicity , Monocytes/cytology , Cell Differentiation , Cells, Cultured , Humans , Macrophages/cytology , Macrophages/physiology , Macrophages/virology , Monocytes/physiology , Monocytes/virology , Virus Replication
10.
J Leukoc Biol ; 76(1): 65-76, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15107461

ABSTRACT

Human cytomegalovirus (HCMV) is a leading cause of morbidity and mortality in immunocompromised hosts. In immunocompetent hosts, HCMV is associated with chronic inflammatory diseases including atherosclerosis. Monocytes and macrophages are proposed to play key roles in HCMV dissemination to host tissue, and their infection provides a biological link between the lifecycle of HCMV and disease pathology. We hypothesize that viral spread occurs via a mechanism in which infected peripheral blood monocytes, which are nonpermissive for viral replication, extravasate into host tissue and subsequently differentiate into permissive macrophages. Supporting this hypothesis, we recently showed that HCMV specifically induced the differentiation of monocytes into macrophages that become permissive for viral replication. To expand our understanding of HCMV pathogenesis, we next examined monocyte activation and migration, the first events in viral pathogenesis. We show here that HCMV up-regulates phosphatidylinositol 3,4,5 triphosphate kinase [PI(3)K] activity and that this increased PI(3)K activity is essential for infected monocyte-transendothelial migration. This increase in migration occurs through the up-regulation of cell motility in a PI(3)K-dependent process. Last, we show that these activated monocytes express a number of inflammatory mediators via PI(3)K signaling. We propose that the up-regulation of monocyte migration and immune mediators by HCMV infection is required for the hematogenous dissemination of the virus and as a consequence, could promote chronic inflammatory diseases associated with HCMV infection.


Subject(s)
Cytomegalovirus Infections/transmission , Macrophage Activation/immunology , Monocytes/cytology , Monocytes/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Blotting, Western , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/immunology , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Endothelium, Vascular/immunology , Enzyme Activation/immunology , Humans , Up-Regulation
11.
J Virol ; 77(22): 12022-32, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14581539

ABSTRACT

C-type lectins such as DC-SIGN and L-SIGN, which bind mannose-enriched carbohydrate modifications of host and pathogen proteins, have been shown to bind glycoproteins of several viruses and facilitate either cis or trans infection. DC-SIGN and L-SIGN are expressed in several early targets of arbovirus infection, including dendritic cells (DCs) and cells of the reticuloendothelial system. In the present study, we show that DC-SIGN and L-SIGN can function as attachment receptors for Sindbis (SB) virus, an arbovirus of the Alphavirus genus. Human monocytic THP-1 cells stably transfected with DC-SIGN or L-SIGN were permissive for SB virus replication, while untransfected controls were essentially nonpermissive. The majority of control THP-1 cells were permissive when attachment and entry steps were eliminated through electroporation of virus transcripts. Infectivity for the DC-SIGN/L-SIGN-expressing cells was largely blocked by yeast mannan, EDTA, or a DC-SIGN/L-SIGN-specific monoclonal antibody. Infection of primary human DCs by SB virus was also dependent upon SIGN expression by similar criteria. Furthermore, production of virus particles in either C6/36 mosquito cells or CHO mammalian cells under conditions that limited complex carbohydrate content greatly increased SB virus binding to and infection of THP-1 cells expressing these lectins. C6/36-derived virus also was much more infectious for primary human DCs than CHO-derived virus. These results suggest that (i) lectin molecules such as DC-SIGN and L-SIGN may represent common attachment receptor molecules for arthropod-borne viruses, (ii) arbovirus particles produced in and delivered by arthropod vectors may preferentially target vertebrate host cells bearing these or similar lectin molecules, and (iii) a cell line has been identified that can productively replicate alphaviruses but is deficient in attachment receptors.


Subject(s)
Cell Adhesion Molecules/physiology , Lectins, C-Type/physiology , Receptors, Cell Surface/physiology , Receptors, Virus/physiology , Sindbis Virus/physiology , Animals , CHO Cells , Cell Line , Cricetinae , Culicidae , Glycoproteins/metabolism , Humans , Virus Replication
12.
J Virol ; 76(18): 9271-83, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12186911

ABSTRACT

Formation of small polykaryons by cell-cell fusion is characteristic of herpes simplex virus (HSV) lesions, but the great majority of viruses isolated from such lesions produce only limited cell fusion in tissue culture. Because of this, HSV laboratory strains that produce extensive cell fusion (syncytium formation) in culture are regarded as variants or mutants. Furthermore, the rarity of clinical isolates able to produce syncytia in culture suggests that extensive cell fusion is deleterious in vivo. Mutations that confer a syncytial phenotype can then be regarded as bypassing a mechanism that normally limits cell fusion. Determination of how these mutations, some of which are in the cytoplasmic tail of glycoprotein B (gB), lead to syncytium formation will likely reveal how fusion is controlled. Here we show the following. (i) Truncation of the cytoplasmic tail of HSV type 2 gB (gB-2) by a minimum of 25 residues or a maximum of 49 residues produces a syncytial phenotype. (ii) Truncation by 20 to 49 residues increases cell fusion when gB-2 is coexpressed with only gD-2, gH-2, and gL-2. (iii) Truncation by 25 or more residues removes a potential endocytosis motif and increases gB-2 cell surface expression. (iv) Mutation of this motif increases gB-2 cell surface expression but does not increase fusogenic activity, whereas mutation of another potential endocytosis motif does not increase surface expression but does increase fusogenic activity. Therefore, syncytial mutations in the cytoplasmic tail of gB-2 do not act by increasing cell surface levels of the protein.


Subject(s)
Giant Cells/physiology , Herpesvirus 2, Human/physiology , Mutation , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Fusion , Cell Line , Herpesvirus 2, Human/genetics , Membrane Fusion , Molecular Sequence Data , Transfection , Viral Envelope Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...