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










Publication year range
2.
Intensive Care Med ; 44(6): 857-867, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29872882

ABSTRACT

PURPOSE: Current guidelines recommend maintaining a mean arterial pressure (MAP) ≥ 65 mmHg in septic patients. However, the relationship between hypotension and major complications in septic patients remains unclear. We, therefore, evaluated associations of MAPs below various thresholds and in-hospital mortality, acute kidney injury (AKI), and myocardial injury. METHODS: We conducted a retrospective analysis using electronic health records from 110 US hospitals. We evaluated septic adults with intensive care unit (ICU) stays ≥ 24 h from 2010 to 2016. Patients were excluded with inadequate blood pressure recordings, poorly documented potential confounding factors, or renal or myocardial histories documented within 6 months of ICU admission. Hypotension exposure was defined by time-weighted average mean arterial pressure (TWA-MAP) and cumulative time below 55, 65, 75, and 85 mmHg thresholds. Multivariable logistic regressions determined the associations between hypotension exposure and in-hospital mortality, AKI, and myocardial injury. RESULTS: In total, 8,782 patients met study criteria. For every one unit increase in TWA-MAP < 65 mmHg, the odds of in-hospital mortality increased 11.4% (95% CI 7.8%, 15.1%, p < 0.001); the odds of AKI increased 7.0% (4.7, 9.5%, p < 0.001); and the odds of myocardial injury increased 4.5% (0.4, 8.7%, p = 0.03). For mortality and AKI, odds progressively increased as thresholds decreased from 85 to 55 mmHg. CONCLUSIONS: Risks for mortality, AKI, and myocardial injury were apparent at 85 mmHg, and for mortality and AKI risk progressively worsened at lower thresholds. Maintaining MAP well above 65 mmHg may be prudent in septic ICU patients.


Subject(s)
Acute Kidney Injury/therapy , Heart Injuries/therapy , Hospital Mortality , Hypotension/therapy , Intensive Care Units/statistics & numerical data , Sepsis/complications , Sepsis/mortality , Acute Kidney Injury/etiology , Acute Kidney Injury/physiopathology , Adult , Aged , Aged, 80 and over , Female , Heart Injuries/etiology , Heart Injuries/physiopathology , Humans , Hypotension/etiology , Hypotension/physiopathology , Male , Middle Aged , Morbidity , Odds Ratio , Retrospective Studies , Risk Factors , United States
3.
J Neurovirol ; 23(1): 152-157, 2017 02.
Article in English | MEDLINE | ID: mdl-27683235

ABSTRACT

Varicella zoster virus (VZV), a human neurotropic alphaherpesvirus, becomes latent after primary infection and reactivates to produce zoster. To study VZV latency and reactivation, human trigeminal ganglia removed within 24 h after death were mechanically dissociated, randomly distributed into six-well tissue culture plates and incubated with reagents to inactivate nerve growth factor (NGF) or phosphoinositide 3-kinase (PI3-kinase) pathways. At 5 days, VZV DNA increased in control and PI3-kinase inhibitor-treated cultures to the same extent, but was significantly more abundant in anti-NGF-treated cultures (p = 0.001). Overall, VZV DNA replication is regulated in part by an NGF pathway that is PI3-kinase-independent.


Subject(s)
DNA Replication , DNA, Viral/genetics , Herpesvirus 3, Human/genetics , Nerve Growth Factor/genetics , Phosphatidylinositol 3-Kinases/genetics , Virus Activation , Virus Replication , Adult , Aged , Antibodies, Neutralizing/pharmacology , Autopsy , DNA, Viral/biosynthesis , Gene Expression Regulation , Herpes Zoster/genetics , Herpes Zoster/metabolism , Herpes Zoster/pathology , Herpes Zoster/virology , Herpesvirus 3, Human/metabolism , Herpesvirus 3, Human/pathogenicity , Host-Pathogen Interactions , Humans , Male , Middle Aged , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factor/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Tissue Culture Techniques , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/virology , Virus Latency
4.
J Virol ; 90(23): 10527-10534, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27630241

ABSTRACT

Varicella-zoster virus (VZV) vasculopathy produces stroke, giant cell arteritis, and granulomatous aortitis, and it develops after virus reactivates from ganglia and spreads transaxonally to arterial adventitia, resulting in persistent inflammation and pathological vascular remodeling. The mechanism(s) by which inflammatory cells persist in VZV-infected arteries is unknown; however, virus-induced dysregulation of programmed death ligand 1 (PD-L1) may play a role. Specifically, PD-L1 can be expressed on virtually all nucleated cells and suppresses the immune system by interacting with the programmed cell death protein receptor 1, found exclusively on immune cells; thus, downregulation of PD-L1 may promote inflammation, as seen in some autoimmune diseases. Both flow cytometry and immunofluorescence analyses to test whether VZV infection of adventitial cells downregulates PD-L1 showed decreased PD-L1 expression in VZV-infected compared to mock-infected human brain vascular adventitial fibroblasts (HBVAFs), perineural cells (HPNCs), and fetal lung fibroblasts (HFLs) at 72 h postinfection. Quantitative RT-PCR analyses showed no change in PD-L1 transcript levels between mock- and VZV-infected cells, indicating a posttranscriptional mechanism for VZV-mediated downregulation of PD-L1. Flow cytometry analyses showed decreased major histocompatibility complex class I (MHC-I) expression in VZV-infected cells and adjacent uninfected cells compared to mock-infected cells. These data suggest that reduced PD-L1 expression in VZV-infected adventitial cells contribute to persistent vascular inflammation observed in virus-infected arteries from patients with VZV vasculopathy, while downregulation of MHC-I prevents viral clearance. IMPORTANCE: Here, we provide the first demonstration that VZV downregulates PD-L1 expression in infected HBVAFs, HPNCs, and HFLs, which, together with the noted VZV-mediated downregulation of MHC-I, might foster persistent inflammation in vessels, leading to pathological vascular remodeling during VZV vasculopathy and persistent inflammation in infected lungs to promote subsequent infection of T cells and hematogenous virus spread. Identification of a potential mechanism by which persistent inflammation in the absence of effective viral clearance occurs in VZV vasculopathy and VZV infection of the lung is a step toward targeted therapy of VZV-induced disease.


Subject(s)
B7-H1 Antigen/metabolism , Herpesvirus 3, Human/pathogenicity , Histocompatibility Antigens Class I/metabolism , Adventitia/blood supply , Adventitia/immunology , Adventitia/virology , B7-H1 Antigen/genetics , Brain/blood supply , Brain/immunology , Brain/virology , Cells, Cultured , Down-Regulation , Fibroblasts/immunology , Fibroblasts/virology , Herpesviridae Infections/etiology , Herpesviridae Infections/immunology , Host-Pathogen Interactions/immunology , Humans , Lung/immunology , Lung/virology , Neuroglia/immunology , Neuroglia/virology
5.
J Neurovirol ; 22(5): 674-682, 2016 10.
Article in English | MEDLINE | ID: mdl-27245593

ABSTRACT

Varicella zoster virus (VZV) is a ubiquitous alphaherpesvirus that establishes latency in ganglionic neurons throughout the neuraxis after primary infection. Here, we show that VZV infection induces a time-dependent significant change in mitochondrial morphology, an important indicator of cellular health, since mitochondria are involved in essential cellular functions. VZV immediate-early protein 63 (IE63) was detected in mitochondria-rich cellular fractions extracted from infected human fetal lung fibroblasts (HFL) by Western blotting. IE63 interacted with cytochrome c oxidase in bacterial 2-hybrid analyses. Confocal microscopy of VZV-infected HFL cells at multiple times after infection revealed the presence of IE63 in the nucleus, mitochondria, and cytoplasm. Our data provide the first evidence that VZV infection induces alterations in mitochondrial morphology, including fragmentation, which may be involved in cellular damage and/or death during virus infection.


Subject(s)
Electron Transport Complex IV/genetics , Fibroblasts/virology , Herpesvirus 3, Human/pathogenicity , Host-Pathogen Interactions , Immediate-Early Proteins/genetics , Mitochondria/virology , Viral Envelope Proteins/genetics , Cell Death/genetics , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Nucleus/virology , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Cytoplasm/virology , Electron Transport Complex IV/metabolism , Fetus , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Herpesvirus 3, Human/growth & development , Humans , Immediate-Early Proteins/metabolism , Lung/cytology , Mitochondria/metabolism , Mitochondria/ultrastructure , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/metabolism
6.
J Neurovirol ; 22(5): 688-694, 2016 10.
Article in English | MEDLINE | ID: mdl-27173396

ABSTRACT

Analysis of the frequency and PCR-quantifiable abundance of herpes simplex virus type 1 (HSV-1) and varicella zoster virus (VZV) DNA in multiple biological replicates of cells from dissociated randomly distributed human trigeminal ganglia (TG) of four subjects revealed an increase in both parameters and in both viruses during 5 days of culture, with no further change by 10 days. Dissociated TG provides a platform to analyze initiation of latent virus DNA replication within 5 days of culture.


Subject(s)
DNA Replication , Herpesvirus 1, Human/physiology , Herpesvirus 3, Human/physiology , Trigeminal Ganglion/virology , Virus Activation , Virus Replication , Adolescent , Adult , Autopsy , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction , Tissue Culture Techniques , Virus Latency
7.
J Neurovirol ; 22(3): 400-2, 2016 06.
Article in English | MEDLINE | ID: mdl-26843382

ABSTRACT

Varicella zoster virus (VZV) becomes latent in ganglionic neurons derived from neural crest cells. Because the adrenal gland also contains medullary chromaffin cells of neural crest origin, we examined human adrenal glands and medullary chromaffin cell tumors (pheochromocytomas) for VZV and herpes simplex virus type 1 (HSV-1). We found VZV, but not HSV-1, DNA in 4/63 (6 %) normal adrenal glands. No VZV transcripts or antigens were detected in the 4 VZV DNA-positive samples. No VZV or HSV-1 DNA was found in 21 pheochromocytomas.


Subject(s)
Adrenal Glands/virology , DNA, Viral/genetics , Herpesvirus 3, Human/genetics , Virus Latency , Adrenal Gland Neoplasms/chemistry , Adrenal Gland Neoplasms/pathology , Adrenal Glands/cytology , Aged, 80 and over , Asymptomatic Diseases , DNA, Viral/isolation & purification , Herpes Simplex , Herpesvirus 1, Human , Herpesvirus 3, Human/isolation & purification , Humans , Male , Middle Aged , Pheochromocytoma/chemistry , Pheochromocytoma/pathology , Polymerase Chain Reaction , Varicella Zoster Virus Infection/virology
8.
J Virol ; 90(3): 1231-43, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26559844

ABSTRACT

UNLABELLED: Regulation of gene transcription in varicella-zoster virus (VZV), a ubiquitous human neurotropic alphaherpesvirus, requires coordinated binding of multiple host and virus proteins onto specific regions of the virus genome. Chromatin immunoprecipitation (ChIP) is widely used to determine the location of specific proteins along a genomic region. Since the size range of sheared virus DNA fragments governs the limit of accurate protein localization, particularly for compact herpesvirus genomes, we used a quantitative PCR (qPCR)-based assay to determine the efficiency of VZV DNA shearing before ChIP, after which the assay was used to determine the relationship between transcript abundance and the occupancy of phosphorylated RNA polymerase II (RNAP) on the gene promoter, body, and terminus of VZV genes 9, 51, and 66. The abundance of VZV gene 9, 51, and 66 transcripts in VZV-infected human fetal lung fibroblasts was determined by reverse transcription-linked quantitative PCR. Our results showed that the C-terminal domain of RNAP is hyperphosphorylated at serine 5 (S5(P)) on VZV genes 9, 51, and 66 independently of transcript abundance and the location within the virus gene at both 1 and 3 days postinfection (dpi). In contrast, phosphorylated serine 2 (S2(P))-modified RNAP was not detected at any virus gene location at 3 dpi and was detected at levels only slightly above background levels at 1 dpi. IMPORTANCE: Regulation of herpesvirus gene transcription is an elaborate choreography between proteins and DNA that is revealed by chromatin immunoprecipitation (ChIP). We used a quantitative PCR-based assay to determine fragment size after DNA shearing, a critical parameter in ChIP assays, and exposed a basic difference in the mechanism of transcription between mammalian cells and VZV. We found that hyperphosphorylation at serine 5 of the C-terminal domain of RNAP along the lengths of VZV genes (the promoter, body, and transcription termination site) was independent of mRNA abundance. In contrast, little to no enrichment of serine 3 phosphorylation of RNAP was detected at these virus gene regions. This is distinct from the findings for RNAP at highly regulated host genes, where RNAP S5(P) occupancy decreased and S2(P) levels increased as the polymerase transited through the gene. Overall, these results suggest that RNAP associates with human and virus transcriptional units through different mechanisms.


Subject(s)
DNA, Viral/chemistry , Herpesvirus 3, Human/physiology , RNA Polymerase II/analysis , Transcription, Genetic , Cells, Cultured , Chromatin Immunoprecipitation , Fibroblasts/virology , Humans , Phosphorylation , Protein Processing, Post-Translational , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
J Gen Virol ; 96(Pt 7): 1581-602, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25794504

ABSTRACT

Herpes simplex virus type 1 (HSV-1; human herpesvirus 1) and varicella-zoster virus (VZV; human herpesvirus 3) are human neurotropic alphaherpesviruses that cause lifelong infections in ganglia. Following primary infection and establishment of latency, HSV-1 reactivation typically results in herpes labialis (cold sores), but can occur frequently elsewhere on the body at the site of primary infection (e.g. whitlow), particularly at the genitals. Rarely, HSV-1 reactivation can cause encephalitis; however, a third of the cases of HSV-1 encephalitis are associated with HSV-1 primary infection. Primary VZV infection causes varicella (chickenpox) following which latent virus may reactivate decades later to produce herpes zoster (shingles), as well as an increasingly recognized number of subacute, acute and chronic neurological conditions. Following primary infection, both viruses establish a latent infection in neuronal cells in human peripheral ganglia. However, the detailed mechanisms of viral latency and reactivation have yet to be unravelled. In both cases latent viral DNA exists in an 'end-less' state where the ends of the virus genome are joined to form structures consistent with unit length episomes and concatemers, from which viral gene transcription is restricted. In latently infected ganglia, the most abundantly detected HSV-1 RNAs are the spliced products originating from the primary latency associated transcript (LAT). This primary LAT is an 8.3 kb unstable transcript from which two stable (1.5 and 2.0 kb) introns are spliced. Transcripts mapping to 12 VZV genes have been detected in human ganglia removed at autopsy; however, it is difficult to ascribe these as transcripts present during latent infection as early-stage virus reactivation may have transpired in the post-mortem time period in the ganglia. Nonetheless, low-level transcription of VZV ORF63 has been repeatedly detected in multiple ganglia removed as close to death as possible. There is increasing evidence that HSV-1 and VZV latency is epigenetically regulated. In vitro models that permit pathway analysis and identification of both epigenetic modulations and global transcriptional mechanisms of HSV-1 and VZV latency hold much promise for our future understanding in this complex area. This review summarizes the molecular biology of HSV-1 and VZV latency and reactivation, and also presents future directions for study.


Subject(s)
Herpesvirus 1, Human/physiology , Herpesvirus 3, Human/physiology , Virus Activation , Virus Latency , Epigenesis, Genetic , Ganglia/virology , Gene Expression Regulation, Viral , Humans , Neurons/virology , Transcription, Genetic
10.
J Virol ; 88(15): 8556-64, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24850726

ABSTRACT

UNLABELLED: The family Arenaviridae includes a number of viruses of public health importance, such as the category A hemorrhagic fever viruses Lassa virus, Junin virus, Machupo virus, Guanarito virus, and Sabia virus. Current chemotherapy for arenavirus infection is limited to the nucleoside analogue ribavirin, which is characterized by considerable toxicity and treatment failure. Using Pichinde virus as a model arenavirus, we attempted to design glycoprotein-derived fusion inhibitors similar to the FDA-approved anti-HIV peptide enfuvirtide. We have identified a GP2-derived peptide, AVP-p, with antiviral activity and no acute cytotoxicity. The 50% inhibitory dose (IC50) for the peptide is 7 µM, with complete inhibition of viral plaque formation at approximately 20 µM, and its antiviral activity is largely sequence dependent. AVP-p demonstrates activity against viruses with the Old and New World arenavirus viral glycoprotein complex but not against enveloped viruses of other families. Unexpectedly, fusion assays reveal that the peptide induces virus-liposome fusion at neutral pH and that the process is strictly glycoprotein mediated. As observed in cryo-electron micrographs, AVP-p treatment causes morphological changes consistent with fusion protein activation in virions, including the disappearance of prefusion glycoprotein spikes and increased particle diameters, and fluorescence microscopy shows reduced binding by peptide-treated virus. Steady-state fluorescence anisotropy measurements suggest that glycoproteins are destabilized by peptide-induced alterations in viral membrane order. We conclude that untimely deployment of fusion machinery by the peptide could render virions less able to engage in on-pathway receptor binding or endosomal fusion. AVP-p may represent a potent, highly specific, novel therapeutic strategy for arenavirus infection. IMPORTANCE: Because the only drug available to combat infection by Lassa virus, a highly pathogenic arenavirus, is toxic and prone to treatment failure, we identified a peptide, AVP-p, derived from the fusion glycoprotein of a nonpathogenic model arenavirus, which demonstrates antiviral activity and no acute cytotoxicity. AVP-p is unique among self-derived inhibitory peptides in that it shows broad, specific activity against pseudoviruses bearing Old and New World arenavirus glycoproteins but not against viruses from other families. Further, the peptide's mechanism of action is highly novel. Biochemical assays and cryo-electron microscopy indicate that AVP-p induces premature activation of viral fusion proteins through membrane perturbance. Peptide treatment, however, does not increase the infectivity of cell-bound virus. We hypothesize that prematurely activated virions are less fit for receptor binding and membrane fusion and that AVP-p may represent a viable therapeutic strategy for arenavirus infection.


Subject(s)
Antiviral Agents/metabolism , Glycoproteins/metabolism , Pichinde virus/drug effects , Pichinde virus/physiology , Virus Internalization/drug effects , Animals , Antiviral Agents/isolation & purification , Cell Line , Cryoelectron Microscopy , Glycoproteins/isolation & purification , Humans , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Pichinde virus/ultrastructure , Viral Plaque Assay , Virion/drug effects , Virion/ultrastructure
11.
Biochim Biophys Acta ; 1838(9): 2180-97, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24780375

ABSTRACT

There are many peptides known that inhibit the entry of enveloped viruses into cells, including one peptide that is successfully being used in the clinic as a drug. In this review, we discuss the discovery, antiviral activity and mechanism of action of such peptides. While peptide entry inhibitors have been discovered by a wide variety of approaches (structure-based, accidental, intentional, rational and brute force) we show here that they share a common physical chemical property: they are at least somewhat hydrophobic and/or amphipathic and have a propensity to interact with membrane interfaces. We propose that this propensity drives a shared mechanism of action for many peptide entry inhibitors, involving direct interactions with viral and cellular membranes, as well as interactions with the complex hydrophobic protein/lipid interfaces that are exposed, at least transiently, during virus-cell fusion. By interacting simultaneously with the membrane interfaces and other critical hydrophobic surfaces, we hypothesize that peptide entry inhibitors can act by changing the physical chemistry of the membranes, and the fusion protein interfaces bridging them, and by doing so interfere with the fusion of cellular and viral membranes. Based on this idea, we propose that an approach that focuses on the interfacial hydrophobicity of putative entry inhibitors could lead to the efficient discovery of novel, broad-spectrum viral entry inhibitors. This article is part of a Special Issue entitled: Interfacially Active Peptides and Proteins. Guest Editors: William C. Wimley and Kalina Hristova.


Subject(s)
Membranes/chemistry , Peptides/chemistry , Viral Envelope Proteins/chemistry , Viral Fusion Proteins/chemistry , Cell Membrane/chemistry , Genome, Viral , Hydrophobic and Hydrophilic Interactions , Membrane Fusion/genetics , Peptides/genetics , Protein Conformation , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics , Virion/genetics
12.
Behav Brain Res ; 208(2): 553-9, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20060021

ABSTRACT

Stress induced by social defeat is a strong modifier of animal anxiety and depression-like phenotypes. Self-grooming is a common rodent behavior, and has an ordered cephalo-caudal progression from licking of the paws to head, body, genitals and tail. Acute stress is known to alter grooming activity levels and disrupt its patterning. Following 15-17 days of chronic social defeat stress, grooming behavior was analyzed in adult male C57BL/6J mice exhibiting either dominant or subordinate behavior. Our study showed that subordinate mice experience higher levels of anxiety and display disorganized patterning of their grooming behaviors, which emerges as a behavioral marker of chronic social stress. These findings indicate that chronic social stress modulates grooming behavior in mice, thus illustrating the importance of grooming phenotypes for neurobehavioral stress research.


Subject(s)
Dominance-Subordination , Grooming/physiology , Stress, Psychological/physiopathology , Animals , Behavior, Animal , Chronic Disease , Exploratory Behavior/physiology , Male , Mice , Mice, Inbred C57BL , Numerical Analysis, Computer-Assisted , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL