Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
bioRxiv ; 2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38645179

ABSTRACT

Human cytomegalovirus (HCMV) infects up to 80% of the world's population. Here, we show that HCMV infection leads to widespread changes in human chromatin accessibility and chromatin looping, with hundreds of thousands of genomic regions affected 48 hours after infection. Integrative analyses reveal HCMV-induced perturbation of Hippo signaling through drastic reduction of TEAD1 transcription factor activity. We confirm extensive concordant loss of TEAD1 binding, active H3K27ac histone marks, and chromatin looping interactions upon infection. Our data position TEAD1 at the top of a hierarchy involving multiple altered important developmental pathways. HCMV infection reduces TEAD1 activity through four distinct mechanisms: closing of TEAD1-bound chromatin, reduction of YAP1 and phosphorylated YAP1 levels, reduction of TEAD1 transcript and protein levels, and alteration of TEAD1 exon-6 usage. Altered TEAD1-based mechanisms are highly enriched at genetic risk loci associated with eye and ear development, providing mechanistic insight into HCMV's established roles in these processes.

2.
Adv Virus Res ; 118: 1-75, 2024.
Article in English | MEDLINE | ID: mdl-38461029

ABSTRACT

G protein coupled receptors (GPCRs) are seven-transmembrane domain proteins that modulate cellular processes in response to external stimuli. These receptors represent the largest family of membrane proteins, and in mammals, their signaling regulates important physiological functions, such as vision, taste, and olfaction. Many organisms, including yeast, slime molds, and viruses encode GPCRs. Cytomegaloviruses (CMVs) are large, betaherpesviruses, that encode viral GPCRs (vGPCRs). Human CMV (HCMV) encodes four vGPCRs, including UL33, UL78, US27, and US28. Each of these vGPCRs, as well as their rodent and primate orthologues, have been investigated for their contributions to viral infection and disease. Herein, we discuss how the CMV vGPCRs function during lytic and latent infection, as well as our understanding of how they impact viral pathogenesis.


Subject(s)
Cytomegalovirus Infections , Receptors, G-Protein-Coupled , Humans , Animals , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Cytomegalovirus/genetics , Cytomegalovirus Infections/metabolism , Mammals/metabolism
3.
Proc Natl Acad Sci U S A ; 120(52): e2313009120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38109533

ABSTRACT

Genetic medicines have the potential to treat various diseases; however, certain ailments including inflammatory diseases and cancer would benefit from control over extracellular localization of therapeutic proteins. A critical gap therefore remains the need to develop and incorporate methodologies that allow for posttranslational control over expression dynamics, localization, and stability of nucleic acid-generated protein therapeutics. To address this, we explored how the body's endogenous machinery controls protein localization through signal peptides (SPs), including how these motifs could be incorporated modularly into therapeutics. SPs serve as a virtual zip code for mRNA transcripts that direct the cell where to send completed proteins within the cell and the body. Utilizing this signaling biology, we incorporated secretory SP sequences upstream of mRNA transcripts coding for reporter, natural, and therapeutic proteins to induce secretion of the proteins into systemic circulation. SP sequences generated secretion of various engineered proteins into the bloodstream following intravenous, intramuscular, and subcutaneous SP mRNA delivery by lipid, polymer, and ionizable phospholipid delivery carriers. SP-engineered etanercept/TNF-α inhibitor proteins demonstrated therapeutic efficacy in an imiquimod-induced psoriasis model by reducing hyperkeratosis and inflammation. An SP-engineered anti-PD-L1 construct mediated mRNA encoded proteins with longer serum half-lives that reduced tumor burden and extended survival in MC38 and B16F10 cancer models. The modular nature of SP platform should enable intracellular and extracellular localization control of various functional proteins for diverse therapeutic applications.


Subject(s)
Dermatitis , Melanoma , Psoriasis , Humans , Animals , Melanoma/drug therapy , Melanoma/genetics , Psoriasis/drug therapy , Psoriasis/genetics , Inflammation/pathology , Protein Sorting Signals , RNA, Messenger/genetics , Disease Models, Animal
4.
Virus Res ; 330: 199112, 2023 06.
Article in English | MEDLINE | ID: mdl-37040820

ABSTRACT

Human cytomegalovirus (HCMV) is ubiquitous in the human population, infecting >70% of people during the course of their lifetime. HCMV DNA and proteins have been detected in glioblastoma (GBM) tumor samples, but whether the virus is a driver of the malignant process or serendipitous passenger is not well understood. Traditionally, HCMV functions in a cytolytic fashion by proceeding through the lytic cycle and spreading viral particles to other cells. Our study focuses on understanding the pattern of HCMV infection and spread within GBM cells using an in vitro model. In cells derived from a GBM biopsy (U373), we found that HCMV does not spread throughout the culture and, in fact, virus-positive cells rapidly decline over time. Interestingly, the viability of the infected GBM cells remained high over the time course, and this was accompanied by a rapid decline in the number of viral genomes over the same time course. The implications of this atypical infection pattern and how this may affect GBM progression is discussed.


Subject(s)
Cytomegalovirus Infections , Glioblastoma , Humans , Cytomegalovirus/genetics , Genome, Viral
5.
Elife ; 112022 09 08.
Article in English | MEDLINE | ID: mdl-36073784

ABSTRACT

The mechanistic details of the tethered agonist mode of activation for the adhesion GPCR ADGRF5/GPR116 have not been completely deciphered. We set out to investigate the physiological importance of autocatalytic cleavage upstream of the agonistic peptide sequence, an event necessary for NTF displacement and subsequent receptor activation. To examine this hypothesis, we characterized tethered agonist-mediated activation of GPR116 in vitro and in vivo. A knock-in mouse expressing a non-cleavable GPR116 mutant phenocopies the pulmonary phenotype of GPR116 knock-out mice, demonstrating that tethered agonist-mediated receptor activation is indispensable for function in vivo. Using site-directed mutagenesis and species-swapping approaches, we identified key conserved amino acids for GPR116 activation in the tethered agonist sequence and in extracellular loops 2/3 (ECL2/3). We further highlight residues in transmembrane 7 (TM7) that mediate stronger signaling in mouse versus human GPR116 and recapitulate these findings in a model supporting tethered agonist:ECL2 interactions for GPR116 activation.


Subject(s)
Pulmonary Surfactants , Amino Acids , Animals , Humans , Mice , Mice, Knockout , Peptides , Pulmonary Surfactants/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
6.
J Virol ; 96(8): e0003122, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35343807

ABSTRACT

Human cytomegalovirus (HCMV) tropism for epithelial cells is determined by the pentameric glycoprotein complex found on the viral envelope. Laboratory-adapted strains, such as AD169, typically develop loss-of-function mutations for the pentamer, thus losing the ability to efficiently initiate lytic replication in epithelial cells. Using our human salivary gland-derived epithelial (hSGE) cell model, we observed that 3 chemically distinct histone deacetylase (HDAC) inhibitors can rescue infection in hSGE cells using pentamer-null strains of HCMV. Additionally, infection in ARPE-19 epithelial cells was rescued in a similar manner. We isolated nuclei from AD169-infected cells, quantified viral genomes by quantitative PCR (qPCR), and discovered that while HDAC inhibitors increased immediate early (IE) gene expression, they did not increase the amount of viral DNA in the nucleus. Using immunofluorescence microscopy, we observed that pentamer-null strains showed punctate patterning of pp71 in proximity to the nucleus of infected cells, while pp71 was localized to the nucleus after infection with pentamer-containing strains. Upon treatment with HDAC inhibitors, these punctae remained perinuclear, while more cells displayed entry into the lytic cycle, noted by increased IE-positive nuclei. Taken together, our data indicate that HCMV pentamer-null viruses are able to infect epithelial cells (albeit less efficiently than pentamer-positive viruses) and traffic to the nucleus but fail to initiate lytic gene expression once there. These studies reveal a novel postentry function of the pentamer in addition to the recognized role of pentamer in mediating entry. IMPORTANCE Human cytomegalovirus has a wide cellular tropism, which is driven by one of its glycoprotein complexes, the pentamer. Laboratory-adapted strains continuously passaged on fibroblasts readily lose pentamer function and thus lose their ability to infect diverse cell types such as epithelial cells. Pentamer has been attributed an entry function during infection, but mechanistic details as to how this is achieved have not been definitely demonstrated. In this study, we investigate how pharmacological rescue of pentamer-null strains during epithelial infection by histone deacetylase inhibitors implicates a novel role for the pentamer downstream of entry. This work expands on potential functions of the pentamer, will drive future studies to understand mechanistically how it affects tropism, and provides a new target for future therapeutics.


Subject(s)
Cytomegalovirus , Epithelial Cells , Histone Deacetylase Inhibitors , Viral Envelope Proteins , Cytomegalovirus/drug effects , Cytomegalovirus/metabolism , Epithelial Cells/virology , Glycoproteins/genetics , Glycoproteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization
7.
Methods Mol Biol ; 2244: 159-197, 2021.
Article in English | MEDLINE | ID: mdl-33555587

ABSTRACT

All of the cytomegaloviruses discovered to date encode two or more genes with significant homology to G protein-coupled receptors (GPCRs). The functions of these cytomegalovirus GPCRs continue to be actively studied and it is clear that they exhibit numerous interesting functions in vitro and in vivo. In this chapter, we review the various methodologies that can be used to examine biochemical aspects of viral GPCR signaling in vitro, as well as examine the biological activity of these viral GPCRs in vitro and in vivo in virus infected cells using recombinant cytomegaloviruses.


Subject(s)
Cell Culture Techniques/methods , Cytomegalovirus/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Cell Line/virology , Cytomegalovirus/metabolism , Humans , Primary Cell Culture/methods , Signal Transduction/genetics , Signal Transduction/physiology
8.
Am J Ind Med ; 63(6): 465-477, 2020 06.
Article in English | MEDLINE | ID: mdl-32270550

ABSTRACT

BACKGROUND: Respirable crystalline silica (RCS) can potentially cause silicosis, lung cancer, and renal failure. The current study estimates the percentages of workers potentially overexposed to concentrations of RCS dust and silicosis proportional mortality rates (PMRs) by industry. METHODS: Occupational Safety and Health Administration compliance inspection sampling data for RCS collected during 1979 to 2015 were used to estimate percentages of workers exposed. The results were used in combination with US Census Bureau estimates to produce industry specific worker population estimates for 2014. Estimates of the numbers and percentages of workers exposed to RCS concentrations at least 1, 2, 5, and 10 times the National Institute for Occupational Safety and Health recommended exposure limit (REL) were calculated by industry using the 2002 North American Industry Classification System. Silicosis PMRs by industry were estimated using National Center for Health Statistics multiple cause of death data. RESULTS: RCS concentrations/workers exposed were highest in the poured concrete foundation and structure contractors; commercial and institutional building construction; and masonry contractors. Approximately 100 000 workers were exposed above the RCS REL, and most (79%) worked in the construction industry. Tile and terrazzo contractors (12%); brick, stone, and related construction merchant wholesalers (10%); masonry contractors (6%) and poured concrete foundation and structure contractors (6%) were the highest percentages of workers potentially overexposed. PMRs were highest for the structural clay product manufacturing and the foundries industries. CONCLUSION: Percentages of workers exposed to RCS varied by industry and in some industries workers are exposed over 10 times the REL. Exposures can be reduced below the REL by implementing the hierarchy of controls.


Subject(s)
Air Pollutants, Occupational/analysis , Industry/statistics & numerical data , Inhalation Exposure/analysis , Occupational Exposure/analysis , Silicon Dioxide/analysis , Silicosis/mortality , Air Pollutants, Occupational/adverse effects , Dust/analysis , Environmental Monitoring/statistics & numerical data , Humans , Inhalation Exposure/adverse effects , Occupational Exposure/adverse effects , Silicosis/etiology , United States/epidemiology , United States Occupational Safety and Health Administration
9.
J Endovasc Ther ; 27(3): 473-480, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32242768

ABSTRACT

Purpose: To evaluate the performance of peripheral intravascular lithotripsy (IVL) in a real-world setting during endovascular treatment of multilevel calcified peripheral artery disease (PAD). Materials and Methods: The Disrupt PAD III Observational Study (ClinicalTrials.gov identifier NCT02923193) is a prospective, nonrandomized, multicenter, single-arm observational study assessing the acute safety and effectiveness of the Shockwave Peripheral IVL System for the treatment of calcified, stenotic lower limb arteries. Patients were eligible if they had claudication or chronic limb-threatening ischemia and moderate or severe arterial calcification. Between November 2017 and August 2018, 200 patients (mean age 72.5±8.7 years; 148 men) were enrolled across 18 sites and followed through hospital discharge. Results: In the 220 target lesions, IVL was more commonly used in combination with other balloon-based technologies (53.8%) and less often with concomitant atherectomy or stenting (19.8% and 29.9%, respectively). There was a 3.4-mm average acute gain at the end of procedure; the final mean residual stenosis was 23.6%. Angiographic complications were rare, with only 2 type D dissections and a single perforation following drug-coated balloon inflation (unrelated to the IVL procedure). There was no abrupt closure, distal embolization, no reflow, or thrombotic event. Conclusion: Use of peripheral IVL to treat severely calcified, stenotic PAD in a real-world study demonstrated low residual stenosis, high acute gain, and a low rate of complications despite the complexity of disease.


Subject(s)
Endovascular Procedures , Intermittent Claudication/therapy , Ischemia/therapy , Lithotripsy , Lower Extremity/blood supply , Peripheral Arterial Disease/therapy , Vascular Calcification/therapy , Aged , Aged, 80 and over , Chronic Disease , Constriction, Pathologic , Endovascular Procedures/adverse effects , Endovascular Procedures/instrumentation , Female , Humans , Intermittent Claudication/diagnostic imaging , Intermittent Claudication/physiopathology , Ischemia/diagnostic imaging , Ischemia/physiopathology , Lithotripsy/adverse effects , Male , Middle Aged , Peripheral Arterial Disease/diagnostic imaging , Peripheral Arterial Disease/physiopathology , Prospective Studies , Time Factors , Treatment Outcome , United States , Vascular Calcification/diagnostic imaging , Vascular Calcification/physiopathology
10.
Proc Natl Acad Sci U S A ; 117(9): 4921-4930, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071223

ABSTRACT

Antibiotic-resistant superbug bacteria represent a global health problem with no imminent solutions. Here we demonstrate that the combination (termed AB569) of acidified nitrite (A-NO2-) and Na2-EDTA (disodium ethylenediaminetetraacetic acid) inhibited all Gram-negative and Gram-positive bacteria tested. AB569 was also efficacious at killing the model organism Pseudomonas aeruginosa in biofilms and in a murine chronic lung infection model. AB569 was not toxic to human cell lines at bactericidal concentrations using a basic viability assay. RNA-Seq analyses upon treatment of P. aeruginosa with AB569 revealed a catastrophic loss of the ability to support core pathways encompassing DNA, RNA, protein, ATP biosynthesis, and iron metabolism. Electrochemical analyses elucidated that AB569 produced more stable SNO proteins, potentially explaining one mechanism of bacterial killing. Our data implicate that AB569 is a safe and effective means to kill pathogenic bacteria, suggesting that simple strategies could be applied with highly advantageous therapeutic/toxicity index ratios to pathogens associated with a myriad of periepithelial infections and related disease scenarios.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Edetic Acid/pharmacology , Sodium Nitrite/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Disease Models, Animal , Down-Regulation , Drug Resistance, Bacterial/drug effects , Edetic Acid/chemistry , Lung Diseases/drug therapy , Lung Diseases/microbiology , Metabolic Networks and Pathways , Mice , Nitrites/chemistry , Nitrites/pharmacology , Pseudomonas aeruginosa/drug effects
11.
J Vis Exp ; (149)2019 07 15.
Article in English | MEDLINE | ID: mdl-31355785

ABSTRACT

The salivary glands are a site of significant interest for researchers interested in multiple aspects of human disease. One goal of researchers is to restore function of glands damaged by radiation therapies or due to pathologies associated with Sjögren's syndrome. A second goal of researchers is to define the mechanisms by which viruses replicate within glandular tissue where they can then gain access to salivary fluids important for horizontal transmission. These goals highlight the need for a robust and accessible in vitro salivary gland model that can be utilized by researchers interested in the above mentioned as well as related research areas. Here we discuss a simple protocol to isolate epithelial cells from human salivary glands and propagate them in vitro. Our protocol can be further optimized to meet the needs of individual studies. Briefly, salivary tissue is mechanically and enzymatically separated to isolate single cells or small clusters of cells. Selection for epithelial cells occurs by plating onto a basement membrane matrix in the presence of media optimized to promote epithelial cell growth. These resulting cultures can be maintained as three-dimensional clusters, termed "salispheres", or grown as a monolayer on treated plastic tissue culture dishes. This protocol results in the outgrowth of a heterogenous population of mainly epithelial cells that can be propagated for 5-8 passages (15-20 population doublings) before undergoing cellular senescence.


Subject(s)
Cell Separation/methods , Epithelial Cells/cytology , Salivary Glands/cytology , Spheroids, Cellular/cytology , Basement Membrane/metabolism , Cell Count , Cell Proliferation , Cells, Cultured , Humans , Submandibular Gland/cytology
12.
mBio ; 10(3)2019 06 25.
Article in English | MEDLINE | ID: mdl-31239384

ABSTRACT

Human cytomegalovirus (HCMV) is a betaherpesvirus that is a significant pathogen within newborn and immunocompromised populations. Morbidity associated with HCMV infection is the consequence of viral dissemination. HCMV has evolved to manipulate the host immune system to enhance viral dissemination and ensure long-term survival within the host. The immunomodulatory protein vCXCL-1, a viral chemokine functioning primarily through the CXCR2 chemokine receptor, is hypothesized to attract CXCR2+ neutrophils to infection sites, aiding viral dissemination. Neutrophils harbor HCMV in vivo; however, the interaction between vCXCL-1 and the neutrophil has not been evaluated in vivo Using the mouse model and mouse cytomegalovirus (MCMV) infection, we show that murine neutrophils harbor and transfer infectious MCMV and that virus replication initiates within this cell type. Utilizing recombinant MCMVs expressing vCXCL-1 from the HCMV strain (Toledo), we demonstrated that vCXCL-1 significantly enhances MCMV dissemination kinetics. Through cellular depletion experiments, we observe that neutrophils impact dissemination but that overall dissemination is largely neutrophil independent. This work adds neutrophils to the list of innate cells (i.e., dendritic and macrophages/monocytes) that contribute to MCMV dissemination but refutes the hypothesis that neutrophils are the primary cell responding to vCXCL-1.IMPORTANCE An adequate in vivo analysis of HCMV's viral chemokine vCXCL-1 has been lacking. Here we generate recombinant MCMVs expressing vCXCL-1 to study vCXCL-1 function in vivo using MCMV as a surrogate. We demonstrate that vCXCL-1 increases MCMV dissemination kinetics for both primary and secondary dissemination. Additionally, we provide evidence, that the murine neutrophil is largely a bystander in the mouse's response to vCXCL-1. We confirm the hypothesis that vCXCL-1 is a HCMV virulence factor. Infection of severely immunocompromised mice with MCMVs expressing vCXCL-1 was lethal in more than 50% of infected animals, while all animals infected with parental virus survived during a 12-day period. This work provides needed insights into vCXCL-1 function in vivo.


Subject(s)
Chemokine CXCL1/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Muromegalovirus/immunology , Neutrophils/virology , Animals , Chemokine CXCL1/genetics , Host-Pathogen Interactions/immunology , Humans , Kinetics , Mice , Mice, Inbred BALB C , Muromegalovirus/pathogenicity , Neutrophils/immunology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , Virulence Factors/immunology , Virus Replication
13.
Proc Natl Acad Sci U S A ; 116(5): 1755-1764, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30647114

ABSTRACT

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that undergoes latency in cells of the hematopoietic compartment, although the mechanisms underlying establishment and maintenance of latency remain elusive. We previously reported that the HCMV-encoded G protein-coupled receptor (GPCR) homolog US28 is required for successful latent infection. We now show that US28 protein (pUS28) provided in trans complements the US28Δ lytic phenotype in myeloid cells, suggesting that sustained US28 expression is necessary for long-term latency. Furthermore, expression of pUS28 at the time of infection represses transcription from the major immediate early promoter (MIEP) within 24 h. However, this repression is only maintained in the presence of continual pUS28 expression provided in trans Our data also reveal that pUS28-mediated signaling attenuates both expression and phosphorylation of cellular fos (c-fos), an AP-1 transcription factor subunit, to repress MIEP-driven transcription. AP-1 binds to the MIEP and promotes lytic replication, and in line with this we find that US28Δ infection results in an increase in AP-1 binding to the MIEP, compared with WT latent infection. Pharmacological inhibition of c-fos represses the MIEP during US28Δ infection to levels similar to those we observe during WT latent infection. Together, our data reveal that US28 is required for both establishment and long-term maintenance of HCMV latency, which is modulated, at least in part, by repressing functional AP-1 binding to the MIEP.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/genetics , Receptors, Chemokine/genetics , Receptors, G-Protein-Coupled/genetics , Viral Proteins/genetics , Virus Latency/genetics , Cell Line , Gene Expression Regulation, Viral/genetics , HEK293 Cells , Humans , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Transcription Factor AP-1/genetics , Virus Replication/genetics
14.
J Virol ; 93(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30404806

ABSTRACT

Various aspects of human cytomegalovirus (HCMV) pathogenesis, including its ability to replicate in specific cells and tissues and the mechanism(s) of horizontal transmission, are not well understood, predominantly because of the strict species specificity exhibited by HCMV. Murine CMV (MCMV), which contains numerous gene segments highly similar to those of HCMV, has been useful for modeling some aspects of CMV pathogenesis; however, it remains essential to build relevant human cell-based systems to investigate how the HCMV counterparts function. The salivary gland epithelium is a site of persistence for both human and murine cytomegaloviruses, and salivary secretions appear to play an important role in horizontal transmission. Therefore, it is important to understand how HCMV is replicating within the glandular epithelial cells so that it might be possible to therapeutically prevent transmission. In the present study, we describe the development of a salivary epithelial model derived from primary human "salispheres." Initial infection of these primary salivary cells with HCMV occurs in a manner similar to that reported for established epithelial lines, in that gH/gL/UL128/UL130/UL131A (pentamer)-positive strains can infect and replicate, while laboratory-adapted pentamer-null strains do not. However, while HCMV enters the lytic phase and produces virus in salivary epithelial cells, it fails to exhibit robust spread throughout the culture and persists in a low percentage of salivary cells. The present study demonstrates the utility of these primary tissue-derived cells for studying HCMV replication in salivary epithelial cells in vitroIMPORTANCE Human cytomegalovirus (HCMV) infects the majority of the world's population, and although it typically establishes a quiescent infection with little to no disease in most individuals, the virus is responsible for a variety of devastating sequelae in immunocompromised adults and in developing fetuses. Therefore, identifying the viral properties essential for replication, spread, and horizontal transmission is an important area of medical science. Our studies use novel human salivary gland-derived cellular models to investigate the molecular details by which HCMV replicates in salivary epithelial cells and provide insight into the mechanisms by which the virus persists in the salivary epithelium, where it gains access to fluids centrally important for horizontal transmission.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/genetics , Epithelium/virology , Fibroblasts/virology , Salivary Glands/virology , Virus Replication , Cells, Cultured , Cytomegalovirus Infections/genetics , Humans , Virus Internalization
15.
Viruses ; 10(8)2018 08 20.
Article in English | MEDLINE | ID: mdl-30127279

ABSTRACT

US28 is one of four G protein coupled receptors (GPCRs) encoded by human cytomegalovirus (HCMV). The US28 protein (pUS28) is a potent signaling molecule that alters a variety of cellular pathways that ultimately alter the host cell environment. This viral GPCR is expressed not only in the context of lytic replication but also during viral latency, highlighting its multifunctional properties. pUS28 is a functional GPCR, and its manipulation of multiple signaling pathways likely impacts HCMV pathogenesis. Herein, we will discuss the impact of pUS28 on both lytic and latent infection, pUS28-mediated signaling and its downstream consequences, and the influence this viral GPCR may have on disease states, including cardiovascular disease and cancer. We will also discuss the potential for and progress towards exploiting pUS28 as a novel therapeutic to combat HCMV.


Subject(s)
Cardiovascular Diseases/virology , Cytomegalovirus Infections/virology , Cytomegalovirus/pathogenicity , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Neoplasms/virology , Receptors, Chemokine/genetics , Viral Proteins/genetics , Cardiovascular Diseases/pathology , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/pathology , Humans , Models, Molecular , Neoplasms/pathology , Protein Structure, Secondary , Receptors, Chemokine/therapeutic use , Signal Transduction , Viral Proteins/therapeutic use , Virus Latency/genetics , Virus Replication/genetics
16.
JCI Insight ; 2(11)2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28570277

ABSTRACT

Pulmonary function is dependent upon the precise regulation of alveolar surfactant. Alterations in pulmonary surfactant concentrations or function impair ventilation and cause tissue injury. Identification of the molecular pathways that sense and regulate endogenous alveolar surfactant concentrations, coupled with the ability to pharmacologically modulate them both positively and negatively, would be a major therapeutic advance for patients with acute and chronic lung diseases caused by disruption of surfactant homeostasis. The orphan adhesion GPCR GPR116 (also known as Adgrf5) is a critical regulator of alveolar surfactant concentrations. Here, we show that human and mouse GPR116 control surfactant secretion and reuptake in alveolar type II (AT2) cells by regulating guanine nucleotide-binding domain α q and 11 (Gq/11) signaling. Synthetic peptides derived from the ectodomain of GPR116 activated Gq/11-dependent inositol phosphate conversion, calcium mobilization, and cortical F-actin stabilization to inhibit surfactant secretion. AT2 cell-specific deletion of Gnaq and Gna11 phenocopied the accumulation of surfactant observed in Gpr116-/- mice. These data provide proof of concept that GPR116 is a plausible therapeutic target to modulate endogenous alveolar surfactant pools to treat pulmonary diseases associated with surfactant dysfunction.

17.
J Gen Virol ; 97(11): 2957-2972, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27638684

ABSTRACT

Cytomegaloviruses (CMVs) produce chemokines (vCXCLs) that have both sequence and functional homology to host chemokines. Assessment of vCXCL-1's role in CMV infection is limited to in vitro and in silico analysis due to CMVs species specificity. In this study, we used the murine CMV (MCMV) mouse model to evaluate the function of vCXCL-1 in vivo. Recombinant MCMVs expressing chimpanzee CMV vCXCL-1 (vCXCL-1CCMV) or host chemokine, mCXCL1, underwent primary dissemination to the popliteal lymph node, spleen and lung similar to the parental MCMV. However, neither of the recombinants expressing chemokines was recovered from the salivary gland (SG) at any time post-infection although viral DNA was detected. This implies that the virus does not grow in the SG or the overexpressed chemokine induces an immune response that leads to suppressed growth. Pointing to immune suppression of virus replication, recombinant viruses were isolated from the SG following infection of immune-ablated mice [i.e. SCID (severe combined immunodeficiency), NSG (non-obese diabetic SCID gamma) or cyclophosphamide treated]. Depletion of neutrophils or NK cells does not rescue the recovery of chemokine-expressing recombinants in the SG. Surprisingly we found that co-infection of parental virus and chemokine-expressing virus leads to the recovery of the recombinants in the SG. We suggest that parental virus reduces the levels of chemokine expression leading to a decrease in inflammatory monocytes and subsequent SG growth. Therefore, aberrant expression of the chemokines induces cells of the innate and adaptive immune system that curtail the growth and dissemination of the recombinants in the SG.


Subject(s)
Chemokines, CXC/immunology , Cytomegalovirus Infections/veterinary , Muromegalovirus/immunology , Salivary Glands/virology , Viral Proteins/immunology , Adaptive Immunity , Animals , Chemokines, CXC/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Host-Pathogen Interactions , Immunity, Innate , Mice , Mice, SCID , Muromegalovirus/genetics , Pan troglodytes , Salivary Glands/immunology , Viral Proteins/genetics
18.
Virology ; 497: 233-243, 2016 10.
Article in English | MEDLINE | ID: mdl-27497185

ABSTRACT

US28 transcripts have been detected in primary monocytes and in THP-1 monocytes infected with HCMV but US28 protein expression has not yet been demonstrated in these cell types. Moreover, the mechanism(s) by which US28 signals and contributes to viral pathogenesis in monocytes remains unclear. Here, we show that US28 protein is robustly expressed in HCMV infected THP-1 monocytes and that US28 can trigger Gαq dependent signaling in THP-1 cells infected with HCMV and in THP-1 cells stably expressing US28. US28 signaling in these cells is dependent on G-protein coupling, but independent of chemokine binding. Importantly, we demonstrate that this US28 signaling is functionally important as it stimulates the adhesion of monocytes to an endothelial monolayer. Our studies, which demonstrate that US28-driven Gαq signaling has profound effects on monocyte biology, suggest that US28 driven phenotypic changes in HCMV infected monocytes may play important roles in HCMV dissemination and/or pathogenesis.


Subject(s)
Cytomegalovirus/physiology , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Monocytes/physiology , Monocytes/virology , Phospholipase C beta/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Viral Proteins/metabolism , Cell Adhesion , Cell Line , Cells, Cultured , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Humans , Receptors, Chemokine/genetics , Viral Proteins/genetics
19.
Cell Rep ; 16(1): 186-200, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27320924

ABSTRACT

Cytomegalovirus (HCMV) contains cholesterol, but how HCMV interacts with host cholesterol metabolism is unknown. We found that, in human fibroblasts, HCMV infection increased the efflux of cellular cholesterol, despite reducing the abundance of ABCA1. Mechanistically, viral protein US28 was acting through CDC42, rearranging actin microfilaments, causing association of actin with lipid rafts, and leading to a dramatic change in the abundance and/or structure of lipid rafts. These changes displaced ABCA1 from the cell surface but created new binding sites for apolipoprotein A-I, resulting in enhanced cholesterol efflux. The changes also reduced the inflammatory response in macrophages. HCMV infection modified the host lipidome profile and expression of several genes and microRNAs involved in cholesterol metabolism. In mice, murine CMV infection elevated plasma triglycerides but did not affect the level and functionality of high-density lipoprotein. Thus, HCMV, through its protein US28, reorganizes lipid rafts and disturbs cell cholesterol metabolism.


Subject(s)
Cholesterol/metabolism , Cytomegalovirus/metabolism , Host-Pathogen Interactions , Membrane Microdomains/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Viral Proteins/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Biological Transport , Cytomegalovirus Infections , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/virology , Humans , Inflammation/pathology , Lipid Metabolism , Male , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred BALB C , RAW 264.7 Cells
20.
Virology ; 483: 83-95, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25965798

ABSTRACT

Human cytomegalovirus (HCMV) resides in a latent form in hematopoietic progenitors and undifferentiated cells within the myeloid lineage. Maturation and differentiation along the myeloid lineage triggers lytic replication. Here, we used peripheral blood monocytes and the monocytic cell line THP-1 to investigate the effects of 1,25-dihydroxyvitamin D3 on HCMV replication. Interestingly, 1,25-dihydroxyvitamin D3 induces lytic replication marked by upregulation of HCMV gene expression and production of infectious virus. Moreover, we demonstrate that the effects of 1,25-dihydroxyvitamin D3 correlate with maturation/differentiation of the monocytes and not by directly stimulating the MIEP. These results are somewhat surprising as 1,25-dihydroxyvitamin D3 typically boosts immunity to bacteria and viruses rather than driving the infectious life cycle as it does for HCMV. Defining the signaling pathways kindled by 1,25-dihydroxyvitamin D3 will lead to a better understanding of the underlying molecular mechanisms that determine the fate of HCMV once it infects cells in the myeloid lineage.


Subject(s)
Calcitriol/metabolism , Cytomegalovirus/drug effects , Cytomegalovirus/growth & development , Monocytes/drug effects , Monocytes/virology , Virus Latency/drug effects , Cells, Cultured , Cytomegalovirus/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...