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










Publication year range
1.
Clin Infect Dis ; 76(8): 1358-1363, 2023 04 17.
Article in English | MEDLINE | ID: mdl-36504336

ABSTRACT

BACKGROUND: In the United States, influenza activity during the 2021-2022 season was modest and sufficient enough to estimate influenza vaccine effectiveness (VE) for the first time since the beginning of the coronavirus disease 2019 pandemic. We estimated influenza VE against laboratory-confirmed outpatient acute illness caused by predominant A(H3N2) viruses. METHODS: Between October 2021 and April 2022, research staff across 7 sites enrolled patients aged ≥6 months seeking outpatient care for acute respiratory illness with cough. Using a test-negative design, we assessed VE against influenza A(H3N2). Due to strong correlation between influenza and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination, participants who tested positive for SARS-CoV-2 were excluded from VE estimations. Estimates were adjusted for site, age, month of illness, race/ethnicity, and general health status. RESULTS: Among 6260 participants, 468 (7%) tested positive for influenza only, including 440 (94%) for A(H3N2). All 206 sequenced A(H3N2) viruses were characterized as belonging to genetic group 3C.2a1b subclade 2a.2, which has antigenic differences from the 2021-2022 season A(H3N2) vaccine component that belongs to clade 3C.2a1b subclade 2a.1. After excluding 1948 SARS-CoV-2-positive patients, 4312 patients were included in analyses of influenza VE; 2463 (57%) were vaccinated against influenza. Effectiveness against A(H3N2) for all ages was 36% (95% confidence interval, 20%-49%) overall. CONCLUSIONS: Influenza vaccination in 2021-2022 provided protection against influenza A(H3N2)-related outpatient visits among young persons.


Subject(s)
COVID-19 , Influenza Vaccines , Influenza, Human , Humans , United States/epidemiology , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Influenza A Virus, H3N2 Subtype , Seasons , Vaccine Efficacy , COVID-19/epidemiology , COVID-19/prevention & control , SARS-CoV-2 , Vaccination , Influenza B virus
2.
PLoS One ; 17(5): e0268038, 2022.
Article in English | MEDLINE | ID: mdl-35503795

ABSTRACT

OBJECTIVES: Studies investigating the association between vitamin D and severity of COVID-19 have mixed results perhaps due to immunoassay assessment of total 25-hydroxyvitamin D (tD) (the sum of 25-hydroxyvitamin-D2 [25-OH-D2] and 25-hydroxyvitamin-D3 [25-OH-D3]). Liquid chromatography tandem mass spectrometry (LC-MS/MS) has high analytical specificity and sensitivity for 25-OH-D2 and 25-OH-D3, and thus enables a more accurate assessment of impact on COVID-19 outcomes. METHODS: We established reference intervals for 25-OH-D3 and tD using LC-MS/MS. 25-OH-D2, 25-OH-D3 and tD were quantitated for 88 COVID-19 positive and 122 COVID-19 negative specimens. Chi-square or Fisher's exact tests were used to test associations in binary variables. T-Tests or Wilcoxon rank sum tests were used for continuous variables. Cox proportional hazards were used to test associations between 25-OH-D3 or tD levels and length of stay (LOS). For mortality and ventilation, logistic regression models were used. RESULTS: COVID-19 patients with deficient (<20 ng/mL) levels of 25-OH-D3 had significantly longer LOS by 15.3 days. COVID-19 P patients with deficient (<20 ng/mL) and insufficient (<30 ng/mL) of tD had significantly longer LOS by 12.1 and 8.2 days, respectively. Patients with insufficient levels of tD had significantly longer LOS by 13.7 days. COVID-19 patients with deficient serum 25-OH-D3 levels had significantly increased risk-adjusted odds of in-hospital mortality (OR [95% CI]: 5.29 [1.53-18.24]); those with insufficient 25-OH-D3 had significantly increased risk for requiring ventilation during hospitalization was found at LCMS insufficient cutoff (OR [95% CI]: 2.75 [1.10-6.90]). CONCLUSIONS: There is an inverse relationship of 25-hydroxyvitamin D levels and hospital LOS for COVID-19 patients. Vitamin D status is a predictor for severity of outcomes. LCMS results are useful for assessing the odds of mortality and the need for ventilation during hospitalization.


Subject(s)
COVID-19 , Tandem Mass Spectrometry , 25-Hydroxyvitamin D 2 , Calcifediol , Chromatography, Liquid/methods , Humans , Tandem Mass Spectrometry/methods , Vitamin D/analogs & derivatives , Vitamins
3.
J Med Virol ; 94(3): 937-944, 2022 03.
Article in English | MEDLINE | ID: mdl-34596257

ABSTRACT

As surges in the COVID-19 pandemic have continued worldwide, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has mutated, spawning several new variants, and impacting, to various degrees, transmission, disease severity, diagnostics, therapeutics, and natural and vaccine-induced immunity. Baylor Scott & White Health has implemented, along with laboratory diagnosis, SARS-CoV-2 sequencing to identify variants in its geographical service area. We analyzed virus sequencing results of specimens collected across Central Texas and found dramatic changes in variant distribution in the first half of 2021. The alpha variant (B 1.1.7) became predominant at week 13 and continued dominance until week 25. A growth rate of 1.20 (R2 = 0.92) for the first 15 weeks was noted and this growth gradually declined to -0.55 (R2 = 0.99) for the final 13 weeks. Currently, B.1.1.7 is being displaced with B.1.617.2 at a 0.58 growth rate (R2 = 0.97). We also investigated vaccine breakthrough cases (VBCs) within our healthcare system and present clinical data on 28 symptomatic patients.


Subject(s)
COVID-19 , Vaccines , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Pandemics , SARS-CoV-2/genetics , Texas/epidemiology
5.
PLoS One ; 15(10): e0240076, 2020.
Article in English | MEDLINE | ID: mdl-33022019

ABSTRACT

Public health emergency of SARS-CoV-2 has facilitated diagnostic testing as a related medical countermeasure against COVID-19 outbreak. Numerous serologic antibody tests have become available through an expedited federal emergency use only process. This paper highlights the analytical characteristic of an ELISA based assay by AnshLabs and three random access immunoassay (RAIA) by DiaSorin, Roche, and Abbott that have been approved for emergency use authorization (EUA), at a tertiary academic center in a low disease-prevalence area. The AnshLabs gave higher estimates of sero-prevalence, over the three RAIA methods. For positive results, AnshLabs had 93.3% and 100% agreement with DiaSorin or Abbott and Roche respectively. For negative results, AnshLabs had 74.3% and 78.3% agreement with DiaSorin and Roche or Abbott respectively. All discrepant samples that were positive by AnshLabs and negative by RAIA tested positive by all-in-one step SARS-CoV-2 Total (COV2T) assay performed on the automated Siemens Advia Centaur XPT analyzer. None of these methods, however, are useful in early diagnosis of SARS-CoV-2.


Subject(s)
Antibodies, Viral/immunology , Betacoronavirus/immunology , Clinical Laboratory Techniques/methods , Coronavirus Infections/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Pneumonia, Viral/diagnosis , Serologic Tests/methods , COVID-19 , COVID-19 Testing , Coronavirus Infections/virology , Diagnostic Tests, Routine , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Nucleocapsid Proteins/immunology , Pandemics , Pneumonia, Viral/virology , Reverse Transcriptase Polymerase Chain Reaction/methods , SARS-CoV-2 , Sensitivity and Specificity , Spike Glycoprotein, Coronavirus/immunology
6.
J Med Virol ; 92(10): 2130-2138, 2020 10.
Article in English | MEDLINE | ID: mdl-32410236

ABSTRACT

Rapid diagnosis and isolation are key to containing the quick spread of a pandemic agent like severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2), which has spread globally since its initial outbreak in Wuhan province in China. SARS-CoV-2 is novel and the effect on typically prevalent seasonal viruses is just becoming apparent. We present our initial data on the prevalence of respiratory viruses in the month of March 2020. This is a retrospective cohort study post launching of SARS-CoV-2 testing at Baylor Scott and White Hospital (BSWH), Temple, Texas. Testing for SARS-CoV-2 was performed by real-time reverse transcription polymerase chain reaction assay and results were shared with State public health officials for immediate interventions. More than 3500 tests were performed during the first 2 weeks of testing for SARS-CoV-2 and identified 168 (4.7%) positive patients. Sixty-two (3.2%) of the 1912 ambulatory patients and 106 (6.3%) of the 1659 emergency department/inpatients tested were positive. The highest rate of infection (6.9%) was seen in patients aged 25 to 34 years, while the lowest rate of infection was seen among patients aged <25 years old (2%). County-specific patient demographic information was shared with respective public health departments for epidemiological interventions. Incidentally, this study showed that there was a significant decrease in the occurrence of seasonal respiratory virus infections, perhaps due to increased epidemiological awareness about SARS-CoV-2 among the general public, as well as the social distancing measures implemented in response to SARS-CoV-2. Data extracted for BSWH from the Centers for Disease Control and Prevention's National Respiratory and Enteric Virus Surveillance System site revealed that Influenza incidence was 8.7% in March 2020, compared with 25% in March 2019. This study was intended to provide an initial experience of dealing with a pandemic and the role of laboratories in crisis management. This study provided SARS-CoV-2 testing data from ambulatory and inpatient population. Epidemiological interventions depend on timely availability of accurate diagnostic tests and throughput capacity of such systems during large outbreaks like SARS-CoV-2.


Subject(s)
COVID-19/epidemiology , Disease Notification/statistics & numerical data , Pandemics , Respiratory Tract Infections/epidemiology , SARS-CoV-2/genetics , Adult , Aged , Aged, 80 and over , COVID-19/diagnosis , COVID-19/transmission , COVID-19/virology , COVID-19 Testing/methods , Epidemiological Monitoring , Female , Humans , Male , Middle Aged , Physical Distancing , Prevalence , Real-Time Polymerase Chain Reaction , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/transmission , Respiratory Tract Infections/virology , Retrospective Studies , Seasons , Texas/epidemiology
7.
J Neuroinflammation ; 13(1): 114, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27207308

ABSTRACT

BACKGROUND: Chemokines produced by reactive glia drive migration of immune cells and previous studies from our laboratory have demonstrated that CD19(+) B cells infiltrate the brain. In this study, in vivo and in vitro experiments investigated the role of reactive glial cells in recruitment and survival of B-lineage cells in response to (murine cytomegalovirus) MCMV infection. METHODS: Flow cytometric analysis was used to assess chemokine receptor expression on brain-infiltrating B cells. Real-time RT-PCR and ELISA were used to measure chemokine levels. Dual-immunohistochemical staining was used to co-localize chemokine production by reactive glia. Primary glial cell cultures and migration assays were used to examine chemokine-mediated recruitment. Astrocyte: B cell co-cultures were used to investigate survival and proliferation. RESULTS: The chemokine receptors CXCR3, CXCR5, CCR5, and CCR7 were detected on CD19(+) cells isolated from the brain during MCMV infection. In particular, CXCR3 was found to be elevated on an increasing number of cells over the time course of infection, and it was the primary chemokine receptor expressed at 60 days post infection Quite different expression kinetics were observed for CXCR5, CCR5, and CCR7, which were elevated on the highest number of cells early during infection and decreased by 14, 30, and 60 days post infection Correspondingly, elevated levels of CXCL9, CXCL10, and CXCL13, as well as CCL5, were found within the brains of infected animals, and only low levels of CCL3 and CCL19 were detected. Differential expression of CXCL9/CXCL10 and CXCL13 between microglia and astrocytes was apparent, and B cells moved towards supernatants from MCMV-infected microglia, but not astrocytes. Pretreatment with neutralizing Abs to CXCL9 and CXCL10 inhibited this migration. In contrast, neutralizing Abs to the ligand of CXCR5 (i.e., CXCL13) did not significantly block chemotaxis. Proliferation of brain-infiltrating B cells was detected at 7 days post infection and persisted through the latest time tested (60 days post infection). Finally, astrocytes produce BAFF (B cell activating factor of the TNF family) and promote proliferation of B cells via cell-to-cell contact. CONCLUSIONS: CXCR3 is the primary chemokine receptor on CD19(+) B cells persisting within the brain, and migration to microglial cell supernatants is mediated through this receptor. Correspondingly, microglial cells produce CXCL9 and CXCL10, but not CXCL13. Reactive astrocytes promote B cell proliferation.


Subject(s)
B-Lymphocytes/pathology , Brain/pathology , Herpesviridae Infections/pathology , Muromegalovirus/pathogenicity , Neuroglia/pathology , Analysis of Variance , Animals , Animals, Newborn , B-Lymphocytes/virology , Brain/virology , Cell Survival , Cells, Cultured , Chemotaxis/physiology , Cytokines/metabolism , Disease Models, Animal , Female , Flow Cytometry , Herpesviridae Infections/virology , Leukocytes/pathology , Mice , Mice, Inbred C57BL , Neuroglia/virology , RNA, Messenger/metabolism
8.
J Clin Diagn Res ; 10(2): DC26-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27042465

ABSTRACT

BACKGROUND: The culture has always been the gold standard test for diagnosis of human brucellosis but the conventional Brucella diagnostic tests viz. serology and culture are often beset with poor specificity & sensitivity respectively. The culture positivity rates for Brucella vary from 92% for bone marrow to 10% for non-blood samples and also dependent on the type of sample. The primary immune-determinant for Brucella species is the cell wall surface lipopolysaccharide, which is antigenically similar to other gram-negative rods. Hence, Brucella serological tests cross react with Escherichia coli 0116 and 0157, Salmonella urbana, Yersinia enterocolitica 0:9, Vibrio cholerae, Xanthomonas maltophilia and Afipia clevellandensis infections, which are common in developing countries also having higher incidence of brucellosis. AIM: The aim of the study was evaluation of conventional serological techniques and PCR for diagnosis of human brucellosis in and around north Karnataka which is endemic for brucellosis and patients often present with elevated base line antibody titers and confounding clinical manifestations. MATERIALS AND METHODS: Blood/serum samples of 400 patients suffering from acute undifferentiated fever (AUF) were subjected to culture, Brucella slide agglutination test (SAT), standard tube agglutination test (STAT coupled with 2 ME) and PCR. RESULTS: Of the 400 AUF patients, anti-Brucella antibodies were detected by SAT and STAT in serum of 35 and 34 patients respectively. IS711 gene for Brucella was identified in 32 patients by PCR. Twenty samples yielded Brucella in culture on biphasic medium with average incubation period of 9 days. All patients having titer of ≥ 160IU / ml in STAT were found positive by PCR also. CONCLUSION: Brucella STAT corroborated well with PCR results in all those cases where antibodies were present at least one dilution above cut-off value of 80 IU/ml. We probably need to raise cut-off titers to ≥160 IU/ml because of endemic region. The SAT was upheld as very good quick, easy to perform and economical screening test for human brucellosis. SAT as rapid screening test and STAT as more definitive test can be very well adopted by laboratories working in resource scarce settings for diagnosis of human brucellosis in absence of PCR even for population with normally elevated antibodies levels due to residing in Brucella endemic areas.

9.
Glia ; 63(11): 1982-1996, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26041050

ABSTRACT

Long-term, persistent central nervous system inflammation is commonly seen following brain infection. Using a murine model of viral encephalitis (murine cytomegalovirus, MCMV) we have previously shown that post-encephalitic brains are maintained in an inflammatory state consisting of glial cell reactivity, retention of brain-infiltrating tissue-resident memory CD8+ T-cells, and long-term persistence of antibody-producing cells of the B-lineage. Here, we report that this neuroinflammation occurs concomitantly with accumulation and retention of immunosuppressive regulatory T-cells (Tregs), and is exacerbated following their ablation. However, the extent to which these Tregs function to control neuroimmune activation following MCMV encephalitis is unknown. In this study, we used Foxp3-diphtheria toxin receptor-GFP (Foxp3-DTR-GFP) transgenic mice, which upon administration of low-dose diphtheria toxin (DTx) results in the specific depletion of Tregs, to investigate their function. We found treatment with DTx during the acute phase of viral brain infection (0-4 dpi) resulted in depletion of Tregs from the brain, exacerbation of encephalitis (i.e., increased presence of CD4+ and CD8+ T-cells), and chronic reactive phenotypes of resident glial cells (i.e., elevated MHC Class II as well as PD-L1 levels, sustained microgliosis, and increased glial fibrillary acidic protein (GFAP) expression on astrocytes) versus untreated, infected animals. This chronic proinflammatory environment was associated with reduced cognitive performance in spatial learning and memory tasks (Barnes Maze) by convalescent animals. These data demonstrate that chronic glial cell activation, unremitting post-encephalitic neuroinflammation, and its associated long-term neurological sequelae in response to viral brain infection are modulated by the immunoregulatory properties of Tregs. GLIA 2015;63:1982-1996.

10.
J Immunol ; 193(12): 6070-80, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25385825

ABSTRACT

Previous studies have demonstrated the existence of a subset of B lymphocytes, regulatory B cells (Bregs), which modulate immune function. In this study, in vivo and in vitro experiments were undertaken to elucidate the role of these Bregs in controlling neuroinflammation following viral brain infection. We used multicolor flow cytometry to phenotype lymphocyte subpopulations infiltrating the brain, along with in vitro cocultures to assess their anti-inflammatory and immunoregulatory roles. This distinctive subset of CD19(+)CD1d(hi)CD5(+) B cells was found to infiltrate the brains of chronically infected animals, reaching highest levels at the latest time point tested (30 d postinfection). B cell-deficient Jh(-/-) mice were found to develop exacerbated neuroimmune responses as measured by enhanced accumulation and/or retention of CD8(+) T cells within the brain, as well as increased levels of microglial activation (MHC class II). Conversely, levels of Foxp3(+) regulatory T cells were found to be significantly lower in Jh(-/-) mice when compared with wild-type (Wt) animals. Further experiments showed that in vitro-generated IL-10-secreting Bregs (B10) were able to inhibit cytokine responses from microglia following stimulation with viral Ags. These in vitro-generated B10 cells were also found to promote proliferation of regulatory T cells in coculture studies. Finally, gain-of-function experiments demonstrated that reconstitution of Wt B cells into Jh(-/-) mice restored neuroimmune responses to levels exhibited by infected Wt mice. Taken together, these results demonstrate that Bregs modulate T lymphocyte as well as microglial cell responses within the infected brain and promote CD4(+)Foxp3(+) T cell proliferation in vitro.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Encephalitis/immunology , Encephalitis/virology , Adoptive Transfer , Animals , B-Lymphocytes, Regulatory/metabolism , Brain/immunology , Brain/pathology , Brain/virology , Disease Models, Animal , Encephalitis/genetics , Encephalitis/metabolism , Female , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Immunophenotyping , Interleukin-10/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Microglia/immunology , Muromegalovirus/genetics , Muromegalovirus/immunology , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
11.
Glia ; 62(10): 1582-94, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24890099

ABSTRACT

Engagement of the programmed death (PD)-1 receptor on activated cells by its ligand (PD-L1) is a mechanism for suppression of activated T-lymphocytes. Microglia, the resident inflammatory cells of the brain, are important for pathogen detection and initiation of innate immunity, however, a novel role for these cells as immune regulators has also emerged. PD-L1 on microglia has been shown to negatively regulate T-cell activation in models of multiple sclerosis and acute viral encephalitis. In this study, we investigated the role of glial cell PD-L1 in controlling encephalitogenic CD8(+) T-lymphocytes, which infiltrate the brain to manage viral infection, but remain to produce chronic neuroinflammation. Using a model of chronic neuroinflammation following murine cytomegalovirus (MCMV)-induced encephalitis, we found that CD8(+) T-cells persisting within the brain expressed PD-1. Conversely, activated microglia expressed PD-L1. In vitro, primary murine microglia, which express low basal levels of PD-L1, upregulated the co-inhibitory ligand on IFN-γ-treatment. Blockade of the PD-1: PD-L1 pathway in microglial: CD8(+) T-cell co-cultures increased T-cell IFN-γ and interleukin (IL)-2 production. We observed a similar phenomenon following blockade of this co-inhibitory pathway in astrocyte: CD8(+) T-cell co-cultures. Using ex vivo cultures of brain leukocytes, including microglia and CD8(+) T-cells, obtained from mice with MCMV-induced chronic neuroinflammation, we found that neutralization of either PD-1 or PD-L1 increased IFN-γ production from virus-specific CD8(+) T-cells stimulated with MCMV IE1168-176 peptide. These data demonstrate that microglia and astrocytes control antiviral T-cell responses and suggest a therapeutic potential of PD1: PD-L1 modulation to manage the deleterious consequences of uncontrolled neuroinflammation.


Subject(s)
Astrocytes/physiology , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/physiology , Encephalitis/physiopathology , Microglia/physiology , Programmed Cell Death 1 Receptor/metabolism , Animals , Brain/physiopathology , Chronic Disease , Coculture Techniques , Cytomegalovirus , Cytomegalovirus Infections , Disease Models, Animal , Female , Interferon-gamma/metabolism , Interleukin-2/metabolism , Leukocytes/physiology , Mice, Inbred BALB C , Neuroimmunomodulation/physiology , Signal Transduction
12.
J Neuroinflammation ; 10: 98, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23902750

ABSTRACT

BACKGROUND: Highly active antiretroviral therapy (HAART) restores inflammatory immune responses in AIDS patients which may unmask previous subclinical infections or paradoxically exacerbate symptoms of opportunistic infections. In resource-poor settings, 25% of patients receiving HAART may develop CNS-related immune reconstitution inflammatory syndrome (IRIS). Here we describe a reliable mouse model to study underlying immunopathological mechanisms of CNS-IRIS. METHODS: Utilizing our HSV brain infection model and mice with MAIDS, we investigated the effect of immune reconstitution on MAIDS mice harboring opportunistic viral brain infection. Using multi-color flow cytometry, we quantitatively measured the cellular infiltrate and microglial activation. RESULTS: Infection with the LP-BM5 retroviral mixture was found to confer susceptibility to herpes simplex virus (HSV)-1 brain infection to normally-resistant C57BL/6 mice. Increased susceptibility to brain infection was due to severe immunodeficiency at 8 wks p.i. and a marked increase in programmed death-1 (PD-1) expression on CD4+ and CD8+ T-cells. Both T-cell loss and opportunistic brain infection were associated with high level PD-1 expression because PD-1-knockout mice infected with LP-BM5 did not exhibit lymphopenia and retained resistance to HSV-1. In addition, HSV-infection of MAIDS mice stimulated peripheral immune cell infiltration into the brain and its ensuing microglial activation. Interestingly, while opportunistic herpes virus brain infection of C57BL/6 MAIDS mice was not itself lethal, when T-cell immunity was reconstituted through adoptive transfer of virus-specific CD3+ T-cells, it resulted in significant mortality among recipients. This immune reconstitution-induced mortality was associated with exacerbated neuroinflammation, as determined by MHC class II expression on resident microglia and elevated levels of Th1 cytokines in the brain. CONCLUSIONS: Taken together, these results indicate development of an immune reconstitution disease within the central nervous system (CNS-IRD). Experimental immune reconstitution disease of the CNS using T-cell repopulation of lymphopenic murine hosts harboring opportunistic brain infections may help elucidate neuroimmunoregulatory networks that produce CNS-IRIS in patients initiating HAART.


Subject(s)
AIDS-Related Opportunistic Infections/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System Viral Diseases/immunology , Herpes Simplex/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , AIDS-Related Opportunistic Infections/mortality , AIDS-Related Opportunistic Infections/pathology , Animals , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Central Nervous System Viral Diseases/mortality , Central Nervous System Viral Diseases/pathology , Herpes Simplex/mortality , Herpes Simplex/pathology , Inflammation/immunology , Inflammation/mortality , Inflammation/pathology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/mortality , Murine Acquired Immunodeficiency Syndrome/pathology
13.
PLoS One ; 7(3): e33143, 2012.
Article in English | MEDLINE | ID: mdl-22412996

ABSTRACT

BACKGROUND: Experimental infection of the mouse brain with murine CMV (MCMV) elicits neuroimmune responses that terminate acute infection while simultaneously preventing extensive bystander damage. Previous studies have determined that CD8(+) T lymphocytes are required to restrict acute, productive MCMV infection within the central nervous system (CNS). In this study, we investigated the contribution of humoral immune responses in control of MCMV brain infection. METHODOLOGY/PRINCIPAL FINDINGS: Utilizing our MCMV brain infection model, we investigated B-lymphocyte-lineage cells and assessed their role in controlling the recovery of reactivated virus from latently infected brain tissue. Brain infiltrating leukocytes were first phenotyped using markers indicative of B-lymphocytes and plasma cells. Results obtained during these studies showed a steady increase in the recruitment of B-lymphocyte-lineage cells into the brain throughout the time-course of viral infection. Further, MCMV-specific antibody secreting cells (ASC) were detected within the infiltrating leukocyte population using an ELISPOT assay. Immunohistochemical studies of brain sections revealed co-localization of CD138(+) cells with either IgG or IgM. Additional immunohistochemical staining for MCMV early antigen 1 (E1, m112-113), a reported marker of viral latency in neurons, confirmed its expression in the brain during latent infection. Finally, using B-cell deficient (Jh(-/-)) mice we demonstrated that B-lymphocytes control recovery of reactivated virus from latently-infected brain tissue. A significantly higher rate of reactivated virus was recovered from the brains of Jh(-/-) mice when compared to Wt animals. CONCLUSION: Taken together, these results demonstrate that MCMV infection triggers accumulation and persistence of B-lymphocyte-lineage cells within the brain, which produce antibodies and play a significant role in controlling reactivated virus.


Subject(s)
Central Nervous System/immunology , Central Nervous System/virology , Herpesviridae Infections/immunology , Immunity, Humoral , Muromegalovirus/immunology , Virus Activation/immunology , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Brain/immunology , Brain/virology , Female , Herpesviridae Infections/virology , Mice , Mice, Inbred BALB C , Mice, Knockout , NIH 3T3 Cells , Neurons/metabolism , Neurons/virology , Plasma Cells/immunology , Virus Latency/immunology
14.
J Neurovirol ; 17(5): 424-37, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21800103

ABSTRACT

Murine cytomegalovirus (MCMV) brain infection stimulates microglial cell-driven proinflammatory chemokine production which precedes the presence of brain-infiltrating systemic immune cells. Here, we show that in response to MCMV brain infection, antigen-specific CD8(+) T cells migrated into the brain and persisted as long-lived memory cells. The role of these persistent T cells in the brain is unclear because most of our understanding of antimicrobial T cell responses comes from analyses of lymphoid tissue. Strikingly, memory T cells isolated from the brain exhibited an effector phenotype and produced IFN-γ upon restimulation with viral peptide. Furthermore, we observed time-dependent and long-term activation of resident microglia, indicated by chronic MHC class II up-regulation and TNF-α production. The immune response in this immunologically restricted site persisted in the absence of active viral replication. Lymphocyte infiltrates were detected until 30 days post-infection (p.i.), with CD8(+) and CD4(+) T cells present at a 3:1 ratio, respectively. We then investigated the role of IFN-γ in chronic microglial activation by using IFN-γ-knockout (GKO) mice. At 30 days p.i., GKO mice demonstrated a similar phenotypic brain infiltrate when compared to wild-type mice (Wt), however, MHC class II expression on microglia isolated from these GKO mice was significantly lower compared to Wt animals. When IFN-γ producing CD8(+) T cells were reconstituted in GKO mice, MHC class II up-regulation on microglial cells was restored. Taken together, these results suggest that MCMV brain infection results in long-term persistence of antigen-specific CD8(+) T cells which produce IFN-γ and drive chronic microglial cell activation. This response was found to be dependent on IFN-γ production by viral Ag-specific T cells during the chronic phase of disease.


Subject(s)
Brain/virology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/pathology , Immunologic Memory , Interferon-gamma/biosynthesis , Microglia/pathology , Animals , Brain/cytology , Brain/immunology , CD4-Positive T-Lymphocytes/immunology , Female , Genes, MHC Class II , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Microglia/immunology , Muromegalovirus/immunology , Muromegalovirus/pathogenicity , Muromegalovirus/physiology , Neutrophils/immunology , Phenotype , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Virus Replication
15.
J Neurovirol ; 17(3): 201-11, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21416394

ABSTRACT

Congenital cytomegalovirus (CMV) infection is the leading cause of sensorineural hearing loss (SNHL) in children. During murine (M)CMV-induced encephalitis, the immune response is important for both the control of viral dissemination and the clearance of virus from the brain. While the importance of CMV-induced SNHL has been described, the mechanisms surrounding its pathogenesis and the role of inflammatory responses remain unclear. This study presents a neonatal mouse model of profound SNHL in which MCMV preferentially infected both cochlear perilymphatic epithelial cells and spiral ganglion neurons. Interestingly, MCMV infection induced cochlear hair cell death by 21 days post-infection, despite a clear lack of direct infection of hair cells and the complete clearance of the virus from the cochlea by 14 dpi. Flow cytometric, immunohistochemical, and quantitative PCR analysis of MCMV-infected cochlea revealed a robust and chronic inflammatory response, including a prolonged increase in reactive oxygen species production by infiltrating macrophages. These data support a pivotal role for inflammation during MCMV-induced SNHL.


Subject(s)
Cochlea/pathology , Cochlea/virology , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/pathology , Epithelial Cells/pathology , Hearing Loss, Sensorineural/etiology , Hearing Loss, Sensorineural/pathology , Neurons/pathology , Animals , Animals, Newborn , Antigens, CD/analysis , Child , Cloning, Molecular , Cochlea/immunology , Cytomegalovirus/genetics , Cytomegalovirus/growth & development , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Disease Models, Animal , Epithelial Cells/virology , Escherichia coli , Evoked Potentials, Auditory, Brain Stem , Female , Flow Cytometry , Hearing Loss, Sensorineural/immunology , Hearing Loss, Sensorineural/virology , Humans , Immunohistochemistry , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Lymphocytes/immunology , Lymphocytes/virology , Macrophages/immunology , Macrophages/virology , Mice , Muromegalovirus/genetics , Muromegalovirus/growth & development , Muromegalovirus/immunology , Neurons/virology , Polymerase Chain Reaction , Recombinant Proteins/genetics , Recombinant Proteins/immunology
16.
PLoS One ; 6(1): e16211, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21249143

ABSTRACT

BACKGROUND: Congenital cytomegalovirus (CMV) brain infection causes serious neuro-developmental sequelae including: mental retardation, cerebral palsy, and sensorineural hearing loss. But, the mechanisms of injury and pathogenesis to the fetal brain are not completely understood. The present study addresses potential pathogenic mechanisms by which this virus injures the CNS using a neonatal mouse model that mirrors congenital brain infection. This investigation focused on, analysis of cell types infected with mouse cytomegalovirus (MCMV) and the pattern of injury to the developing brain. METHODOLOGY/PRINCIPAL FINDINGS: We used our MCMV infection model and a multi-color flow cytometry approach to quantify the effect of viral infection on the developing brain, identifying specific target cells and the consequent effect on neurogenesis. In this study, we show that neural stem cells (NSCs) and neuronal precursor cells are the principal target cells for MCMV in the developing brain. In addition, viral infection was demonstrated to cause a loss of NSCs expressing CD133 and nestin. We also showed that infection of neonates leads to subsequent abnormal brain development as indicated by loss of CD24(hi) cells that incorporated BrdU. This neonatal brain infection was also associated with altered expression of Oct4, a multipotency marker; as well as down regulation of the neurotrophins BDNF and NT3, which are essential to regulate the birth and differentiation of neurons during normal brain development. Finally, we report decreased expression of doublecortin, a marker to identify young neurons, following viral brain infection. CONCLUSIONS: MCMV brain infection of newborn mice causes significant loss of NSCs, decreased proliferation of neuronal precursor cells, and marked loss of young neurons.


Subject(s)
Brain/virology , Herpesviridae Infections , Muromegalovirus , Neural Stem Cells/virology , Neurogenesis , Animals , Animals, Newborn , Biomarkers/analysis , Brain/growth & development , Brain/pathology , Brain Diseases/metabolism , Brain Diseases/pathology , Brain Diseases/virology , Cell Proliferation , Flow Cytometry , Mice
17.
Vet Immunol Immunopathol ; 140(1-2): 30-6, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21146877

ABSTRACT

Commercial turkey eggs, free of antibodies to avian metapneumovirus subtype C (aMPV/C), were inoculated with aMPV/C at embryonation day (ED) 24. There was no detectable effect of virus inoculation on the hatchability of eggs. At 4 days post inoculation (DPI) (the day of hatch (ED 28)) and 9 DPI (5 days after hatch), virus replication was detected by quantitative RT-PCR in the turbinate, trachea and lung but not in the thymus or spleen. Mild histological lesions characterized by lymphoid cell infiltration were evident in the turbinate mucosa. Virus exposure inhibited the mitogenic response of splenocytes and thymocytes and upregulated gene expression of IFN-γ and IL-10 in the turbinate tissue. Turkeys hatching from virus-exposed eggs had aMPV/C-specific IgG in the serum and the lachrymal fluid. At 3 week of age, in ovo immunized turkeys were protected against a challenge with pathogenic aMPV/C.


Subject(s)
Metapneumovirus/immunology , Paramyxoviridae Infections/veterinary , Poultry Diseases/immunology , Vaccination/veterinary , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Embryo, Nonmammalian/immunology , Enzyme-Linked Immunosorbent Assay/veterinary , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-18/biosynthesis , Metapneumovirus/isolation & purification , Mitogens/immunology , Paramyxoviridae Infections/blood , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Poultry Diseases/blood , Poultry Diseases/virology , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Turkeys
18.
J Neuroimmunol ; 227(1-2): 101-10, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20655600

ABSTRACT

Wild-type mice control murine cytomegalovirus (MCMV) brain infection, but identical infection is lethal to animals deficient in interleukin (IL)-10. Here, we report that MCMV-infected IL-10 knockout (KO) mice displayed a marked increase in neutrophil infiltration into the infected, IL-10-deficient brain when compared to wild-type animals. Enhanced microglial cell activation, determined by MHC class II up-regulation, overexpression of CXCL2, and elevated P-selectin mRNA levels were observed. In vivo blocking of CXCL2 attenuated neutrophil infiltration and significantly improved the outcome of infection. Collectively, these data indicate that the absence of IL-10 results in pathologic neutrophil infiltration into MCMV-infected brains.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Encephalitis, Viral/immunology , Encephalitis, Viral/pathology , Interleukin-10/deficiency , Muromegalovirus/immunology , Neutrophil Infiltration/immunology , Animals , Cells, Cultured , Cytomegalovirus Infections/genetics , Encephalitis, Viral/genetics , Female , Humans , Interleukin-10/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , NIH 3T3 Cells , Neutrophil Infiltration/genetics , Rats
19.
J Neurovirol ; 15(4): 334-42, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19626525

ABSTRACT

Interleukin (IL)-10 deficiency results in highly elevated levels of interferon (IFN)-gamma, as well as the IFN-gamma-inducible chemokines CXCL9 and CXCL10 within murine cytomegalovirus (MCMV)-infected brains. To test the hypothesis that these elevated chemokine levels would result in enhanced brain infiltration, we compared immune cell infiltration in response to MCMV brain infection between wild-type and IL-10 knockout (KO) mice. Longitudinal analysis following adoptive transfer of cells from beta-actin-luciferase transgenic wild-type mice showed maximal brain infiltration by peripheral immune cells occurred at 5 days post infection. Although the overall percentage of CD45(hi) cells infiltrating the brain was not altered by IL-10 deficiency, paradoxically, despite elevated chemokine levels, reduced T lymphocyte (CD8+) and natural killer (NK) (CD49b+) cell infiltration into the brain was observed in IL-10-deficient animals. This decreased lymphocyte infiltration was associated with elevated levels of the lymph node homing receptor L-selectin/CD62L on CD8+ T cells. Lymph node cells obtained from MCMV-infected mice deficient in IL-10 also displayed reduced migration towards CXCL10 when compared to wild-type animals. Taken together, these data show that despite elevated chemokine levels, absence of IL-10 results in reduced lymphocyte infiltration into MCMV-infected brains.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Encephalitis/immunology , Interleukin-10/deficiency , Killer Cells, Natural/metabolism , Muromegalovirus/immunology , Animals , Animals, Genetically Modified , Brain/immunology , Brain/virology , CD8-Positive T-Lymphocytes/cytology , Chemokine CXCL10 , Encephalitis/virology , Female , L-Selectin/metabolism , Luciferases , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Spleen/cytology
20.
J Med Microbiol ; 56(Pt 2): 283-285, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17244815

ABSTRACT

Human brucellosis, a zoonotic disease, is endemic in the Belgaum district, Karnataka, India. A male patient presented with a generalized itchy rash. Blood was sent for venereal disease research laboratory testing. Screening was carried out for Brucella antibodies following hospital policy and diagnosis was confirmed by PCR.


Subject(s)
Brucella melitensis/isolation & purification , Brucellosis/diagnosis , DNA, Bacterial/analysis , Polymerase Chain Reaction , Skin Diseases, Bacterial/microbiology , Animals , Antibodies, Bacterial , Brucella melitensis/genetics , Brucellosis/microbiology , Brucellosis/physiopathology , DNA, Bacterial/genetics , Endemic Diseases , Exanthema , Humans , India , Male , Middle Aged , Pruritus , Skin Diseases, Bacterial/physiopathology , Zoonoses
SELECTION OF CITATIONS
SEARCH DETAIL
...