Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Genes Brain Behav ; 19(8): e12690, 2020 11.
Article in English | MEDLINE | ID: mdl-32741046

ABSTRACT

Dyskinesia induced by long-term L-Dopa (LID) therapy in Parkinson disease is associated with altered striatal function whose molecular bases remain unclear. Here, a transcriptomic approach was applied for comprehensive analysis of distinctively regulated genes in striatal tissue, their specific pathways, and functional- and disease-associated networks in a rodent model of LID. This approach has identified transforming growth factor beta type 1 (TGFß1) as a highly upregulated gene in dyskinetic animals. TGFß1 pathway is a top aberrantly regulated pathway in the striatum following LID development based on differentially expressed genes (> 1.5 fold change and P < 0.05). The induction of TGFß1 pathway specific genes, TGFß1, INHBA, AMHR2 and PMEPA1 was also associated with regulation of NPTX2, PDP1, SCG2, SYNPR, TAC1, TH, TNNT1 genes. Transcriptional network and upstream regulator analyses have identified AKT-centered functional and ERK-centered disease networks revealing the association of TGFß1, IL-1ß and TNFα with LID development. Therefore, results support that TGFß1 pathway is a major contributor to the pathogenic mechanisms of LID.


Subject(s)
Dyskinesia, Drug-Induced/metabolism , Signal Transduction , Transcriptome , Transforming Growth Factor beta1/metabolism , Animals , Antiparkinson Agents/toxicity , Brain/metabolism , Dyskinesia, Drug-Induced/genetics , Gene Regulatory Networks , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Levodopa/toxicity , Male , Rats , Rats, Sprague-Dawley , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Tachykinins/genetics , Tachykinins/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta1/genetics , Up-Regulation
2.
Antiviral Res ; 180: 104849, 2020 08.
Article in English | MEDLINE | ID: mdl-32553844

ABSTRACT

During the last decade multiple mumps outbreaks have occurred in the U.S. despite high two dose MMR coverage with most cases detected among two dose MMR vaccine recipients. Waning immunity, the evolution of wild-type virus strains, and settings with intense exposure have contributed to the resurgence of mumps. Typically, mumps virus infections resolve without serious clinical sequelae; however, serious complications may occur among unvaccinated or severely immunocompromised individuals. Favipiravir (T-705) has been shown to have in vitro anti-viral activity against a broad range of positive and negative strand RNA viruses. Here, we demonstrate that T-705 inhibits the growth of wildtype and vaccine strains of mumps virus in vitro at low micro-molar concentrations (EC50 8-10µM). We did not observe the development of resistance after five subsequent passages at low concentrations of drug. Both viral RNA and protein synthesis were selectively reduced compared to host mRNA and protein synthesis. Antiviral treatment options for mumps virus infection may be valuable, especially for areas with a high disease burden or for cases with severe complications. These results presented here suggest that further studies are warranted.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Mumps virus/drug effects , Pyrazines/pharmacology , Virus Replication/drug effects , A549 Cells , Animals , Chlorocebus aethiops , Humans , RNA, Viral/antagonists & inhibitors , Vero Cells
4.
Vaccine ; 38(9): 2172-2182, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32008879

ABSTRACT

Ever since its development in the 1930's, the live-attenuated Yellow Fever virus vaccine YF-17D has been highly effective. Despite the increasing knowledge on the immune biology of the YF-17D vaccine, most studies have focused only on a few types of immune cells and pathways or mainly on the primary adaptive immune response to YF-17D vaccination. Here, we examined humoral, innate and adaptive cellular responses in a longitudinal YF-17D vaccination study in Switzerland, comparing both primary and booster vaccination. In contrast to the strong innate and adaptive immune response to the primary vaccination, we find that the response to boosting is much reduced. Our data show an inverse association of neutralizing antibodies at baseline with vaccine virus replication and with the immune response upon boosting. These results suggest that booster vaccination may not have major immunological effects when neutralizing antibodies are present. Importantly, our study population was healthy adults in a non-endemic country and ultimately booster vaccine requirement must be assessed based on additional epidemiological and public health considerations in endemic areas.


Subject(s)
Antibodies, Viral/immunology , Immunization, Secondary , Yellow Fever Vaccine/administration & dosage , Yellow Fever/immunology , Adult , Antibodies, Neutralizing/immunology , Humans , Immunity, Cellular , Immunity, Humoral , Immunity, Innate , Switzerland , Yellow Fever/prevention & control
5.
Mil Med ; 184(7-8): e296-e300, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30951145

ABSTRACT

INTRODUCTION: One challenge clinicians face is determining when a military Service Member (SM) can return to duty after an injury that affects the postural control. The gold standard to measure postural control is the Sensory Organization Test (SOT). This test measures the amount of sway present in an individual's static stance that may be used to examine range of function and monitor recovery from injury. Normative values currently available were developed using a sample of clinically normal adults from the general population (i.e., civilian non-aviator). Previous research suggests that these values should not be used as a comparative cohort for high-performing populations in the military. However, normative values, specific to military SMs, do not exist. The aim of this study was to develop a normative clinical database for functional balance (i.e., the SOT) for military-trained aviators, an occupational specialty that may consist of high performers. MATERIALS AND METHODS: Forty-three U.S. Army trained aviators, between 23 and 40 years old with medical clearance for flight operations from the Fort Rucker, Alabama area community consented and participated in this study. The SOT was delivered using the NeuroCom SMART EquiTest Clinical Research System with the Data Acquisition Toolkit (version 9.3). RESULTS: A statistically significant (p < 0.01) difference between the study cohort of Army-trained aviators and the publically available general civilian normative values was found for the more challenging conditions, in which the force plate was not fixed (i.e., conditions four through six). The study cohort of Army-trained aviators were found to have a higher equilibrium score in each of these three conditions. Similarly, a significant difference (p < 0.01) between the two cohorts was found on the visual and vestibular sensory analysis ratios, and the visual preference scores (i.e., greater reliance upon visual information in the maintenance of balance). The study cohort were found to have a higher ratios (i.e., greater dependence upon these sensory cues) in each of these conditions. CONCLUSION: Army-trained aviators are high-functioning performers whose SOT scores differ from that of the general civilian population, particularly for the more challenging test conditions. New normative values were developed from this study population. Use of the developed normative values could be used as a comparative cohort in screening aviators who are recovering from injuries that affect postural stability.


Subject(s)
Military Personnel/statistics & numerical data , Pilots/classification , Postural Balance/physiology , Adult , Alabama/epidemiology , Female , Gait Disorders, Neurologic/complications , Gait Disorders, Neurologic/epidemiology , Humans , Male , Military Personnel/classification
6.
Pathog Immun ; 4(1): 1-20, 2019.
Article in English | MEDLINE | ID: mdl-30993250

ABSTRACT

BACKGROUND: Vaccination with the Merck human adenovirus serotype-5 (HAdV-5) vectored HIV-1 subtype B gag/pol/nef vaccine was unexpectedly associated with enhanced susceptibility to HIV-1 infection in uncircumcised HAdV-5 seropositive men. It has been hypothesized that vaccination may have resulted in activated CD4+ T lymphocytes trafficking to mucosal sites thereby increasing targets for HIV infection. We have previously shown that AdV-vector vacci-nation in rhesus macaques resulted in an increase in the frequency of activated mucosal CD4+ T cells. However, whether this increase in activation is sufficient to increase susceptibility to HIV/SIV infection is unclear. METHODS: To examine this scenario, we developed a preliminary, proof-of-concept vaccination-challenge model in order to examine vaccine-induced SIV susceptibility in rhesus macaques. Rhesus macaques (n = 10/group) were vaccinated with a simian AdV-7 (SAdV-7)-vector encoding an irrelevant insert (SARS spike) and challenged 5 weeks post-prime in an escalating dosing regimen starting with sub-infectious doses (1:10,000 or 2TCID50) of SIVmac251. RESULTS: In contrast to our previous study, the SAdV-7 vaccine regimen did not induce detectable mucosal CD4+ T cell activation at the time points assessed in animals obtained from a different vendor and housed in a different facility. Within the power of the study, we did not observe significantly increased SIV acquisition in SAdV-7-vaccinated (5/10) versus placebo-vaccinated (3/10) macaques after repeated low-dose intra-rectal SIVmac251 challenge (P < 0.2). CONCLUSIONS: These results lay groundwork for future experiments to assess vaccine-induced SIV susceptibility in rhesus macaques. Further larger-scale studies are necessary to confirm the AdV-vector vaccination associated trend towards increased SIV/HIV acquisition and clarify associated mechanisms.

8.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29997216

ABSTRACT

Worldwide, nearly two million children are infected with human immunodeficiency virus (HIV), with breastfeeding accounting for the majority of contemporary HIV transmissions. Antiretroviral therapy (ART) has reduced HIV-related morbidity and mortality but is not curative. The main barrier to a cure is persistence of latent HIV in long-lived reservoirs. However, our understanding of the cellular and anatomic sources of the HIV reservoir during infancy and childhood is limited. Here, we developed a pediatric model of ART suppression in orally simian immunodeficiency virus (SIV)-infected rhesus macaque (RM) infants, with measurement of virus persistence in blood and tissues after 6 to 9 months of ART. Cross-sectional analyses were conducted to compare SIV RNA and DNA levels in adult and infant RMs naive to treatment and on ART. We demonstrate efficient viral suppression following ART initiation in SIV-infected RM infants with sustained undetectable plasma viral loads in the setting of heterogeneous penetration of ART into lymphoid and gastrointestinal tissues and low drug levels in the brain. We further show reduction in SIV RNA and DNA on ART in lymphoid tissues of both infant and adult RMs but stable (albeit low) levels of SIV RNA and DNA in the brains of viremic and ART-suppressed infants. Finally, we report a large contribution of naive CD4+ T cells to the total CD4 reservoir of SIV in blood and lymph nodes of ART-suppressed RM infants that differs from what we show in adults. These results reveal important aspects of HIV/SIV persistence in infants and provide insight into strategic targets for cure interventions in a pediatric population.IMPORTANCE While antiretroviral therapy (ART) can reduce HIV replication, the virus cannot be eradicated from an infected individual, and our incomplete understanding of HIV persistence in reservoirs greatly complicates the generation of a cure for HIV infection. Given the immaturity of the infant immune system, it is critically important to study HIV reservoirs specifically in this population. Here, we established a pediatric animal model to simulate breastfeeding transmission and study SIV reservoirs in rhesus macaque (RM) infants. Our study demonstrates that ART can be safely administered to infant RMs for prolonged periods and that it efficiently controls viral replication in this model. SIV persistence was shown in blood and tissues, with similar anatomic distributions of SIV reservoirs in infant and adult RMs. However, in the peripheral blood and lymph nodes, a greater contribution of the naive CD4+ T cells to the SIV reservoir was observed in infants than in adults.


Subject(s)
Antiretroviral Therapy, Highly Active/adverse effects , Infectious Disease Transmission, Vertical/veterinary , Simian Acquired Immunodeficiency Syndrome/blood , Simian Immunodeficiency Virus/isolation & purification , Viral Load/drug effects , Animals , CD4 Lymphocyte Count , Cross-Sectional Studies , Disease Reservoirs , Lymph Nodes/immunology , Lymph Nodes/virology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology
9.
J Virol ; 92(9)2018 05 01.
Article in English | MEDLINE | ID: mdl-29467313

ABSTRACT

Pathogenic human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) infection of humans and rhesus macaques (RMs) induces persistently high production of type I interferon (IFN-I), which is thought to contribute to disease progression. To elucidate the specific role of interferon alpha (IFN-α) in SIV pathogenesis, 12 RMs were treated prior to intravenous (i.v.) SIVmac239 infection with a high or a low dose of an antibody (AGS-009) that neutralizes most IFN-α subtypes and were compared with six mock-infused, SIV-infected controls. Plasma viremia was measured postinfection to assess the effect of IFN-α blockade on virus replication, and peripheral blood and lymphoid tissue samples were analyzed by immunophenotypic staining. Consistent with the known antiviral effect of IFN-I, high-dose AGS-009 treatment induced a modest increase in acute-phase viral loads versus controls. Four out of 6 RMs receiving a high dose of AGS-009 also experienced an early decline in CD4+ T cell counts that was associated with progression to AIDS. Interestingly, 50% of the animals treated with AGS-009 (6/12) developed AIDS within 1 year of infection compared with 17% (1/6) of untreated controls. Finally, blockade of IFN-α decreased the levels of activated CD4+ and CD8+ T cells, as well as B cells, as measured by PD-1 and/or Ki67 expression. The lower levels of activated lymphocytes in IFN-α-blockaded animals supports the hypothesis that IFN-α signaling contributes to lymphocyte activation during SIV infection and suggests that this signaling pathway is involved in controlling virus replication during acute infection. The potential anti-inflammatory effect of IFN-α blockade should be explored as a strategy to reduce immune activation in HIV-infected individuals.IMPORTANCE Interferon alpha (IFN-α) is a member of a family of molecules (type I interferons) that prevent or limit virus infections in mammals. However, IFN-α production may contribute to the chronic immune activation that is thought to be the primary cause of immune decline and AIDS in HIV-infected patients. The study presented here attempts to understand the contribution of IFN-α to the natural history and progression of SIV infection of rhesus macaques, the primary nonhuman primate model system for testing hypotheses about HIV infection in humans. Here, we show that blockade of IFN-α action promotes lower chronic immune activation but higher early viral loads, with a trend toward faster disease progression. This study has significant implications for new treatments designed to impact the type I interferon system.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-alpha/antagonists & inhibitors , Lymphocyte Activation/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Animals , Antibodies, Monoclonal/immunology , B-Lymphocytes/immunology , CD4 Lymphocyte Count , Interferon-alpha/immunology , Ki-67 Antigen/biosynthesis , Killer Cells, Natural/immunology , Macaca mulatta , Programmed Cell Death 1 Receptor/biosynthesis , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Viral Load/drug effects , Virus Replication/immunology
10.
J Immunol ; 197(9): 3586-3596, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27683750

ABSTRACT

We tested, in rhesus macaques, the effects of a 500-fold range of an admixed recombinant modified vaccinia Ankara (MVA) expressing rhesus GM-CSF (MVA/GM-CSF) on the immunogenicity and protection elicited by an MVA/SIV macaque 239 vaccine. High doses of MVA/GM-CSF did not affect the levels of systemic envelope (Env)-specific Ab, but it did decrease the expression of the gut-homing receptor α4ß7 on plasmacytoid dendritic cells (p < 0.01) and the magnitudes of Env-specific IgA (p = 0.01) and IgG (p < 0.05) in rectal secretions. The protective effect of the vaccine was evaluated using 12 weekly rectal challenges in rhesus macaques subgrouped by tripartite motif-containing protein 5α (TRIM5α) genotypes that are restrictive or permissive for infection by the challenge virus SIVsmE660. Eight of nine TRIM5α-restrictive animals receiving no or the lowest dose (1 × 105 PFU) of MVA/GM-CSF resisted all 12 challenges. In the comparable TRIM5α-permissive group, only 1 of 12 animals resisted all 12 challenges. In the TRIM5α-restrictive animals, but not in the TRIM5α-permissive animals, the number of challenges to infection directly correlated with the magnitudes of Env-specific rectal IgG (r = +0.6) and IgA (r = +0.6), the avidity of Env-specific serum IgG (r = +0.5), and Ab dependent cell-mediated virus inhibition (r = +0.6). Titers of neutralizing Ab did not correlate with protection. We conclude that 1) protection elicited by MVA/SIVmac239 is strongly dependent on the presence of TRIM5α restriction, 2) nonneutralizing Ab responses contribute to protection against SIVsmE660 in TRIM5α-restrictive animals, and 3) high doses of codelivered MVA/GM-CSF inhibit mucosal Ab responses and the protection elicited by MVA expressing noninfectious SIV macaque 239 virus-like particles.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Rectum/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Antibody Formation , Genotype , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Immunoglobulin G/metabolism , Macaca mulatta , Proteins/genetics , Simian Immunodeficiency Virus/immunology , Ubiquitin-Protein Ligases , Vaccines, DNA , Vaccinia/immunology , Viral Envelope Proteins/immunology
11.
Immunity ; 45(3): 656-668, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27653601

ABSTRACT

Infection with HIV persists despite suppressive antiretroviral therapy (ART), and treatment interruption results in rapid viral rebound. Antibody-mediated CD8(+) lymphocyte depletion in simian immunodeficiency virus (SIV)-infected rhesus macaques (RMs) shows that these cells contribute to viral control in untreated animals. However, the contribution of CD8(+) lymphocytes to maintaining viral suppression under ART remains unknown. Here, we have shown that in SIV-infected RMs treated with short-term (i.e., 8-32 week) ART, depletion of CD8(+) lymphocytes resulted in increased plasma viremia in all animals and that repopulation of CD8(+) T cells was associated with prompt reestablishment of virus control. Although the number of SIV-DNA-positive cells remained unchanged after CD8 depletion and reconstitution, the frequency of SIV-infected CD4(+) T cells before depletion positively correlated with both the peak and area under the curve of viremia after depletion. These results suggest a role for CD8(+) T cells in controlling viral production during ART, thus providing a rationale for exploring immunotherapeutic approaches in ART-treated HIV-infected individuals.


Subject(s)
Anti-Retroviral Agents/pharmacology , CD8-Positive T-Lymphocytes/immunology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Viral/immunology , Antiretroviral Therapy, Highly Active/methods , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Female , Lymphocyte Depletion/methods , Macaca mulatta , Male , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Viral Load/drug effects , Viral Load/immunology , Viremia/drug therapy , Viremia/immunology , Viremia/virology , Virus Replication/drug effects , Virus Replication/immunology
12.
J Virol ; 90(16): 7541-7551, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27279614

ABSTRACT

UNLABELLED: Simian immunodeficiency virus (SIV)-infected sooty mangabeys (SMs) do not develop AIDS despite high levels of viremia. Key factors involved in the benign course of SIV infection in SMs are the absence of chronic immune activation and low levels of infection of CD4(+) central memory (TCM) and stem cell memory (TSCM) T cells. To better understand the role of virus replication in determining the main features of SIV infection in SMs, we treated 12 SMs with a potent antiretroviral therapy (ART) regimen for 2 to 12 months. We observed that ART suppressed viremia to <60 copies/ml of plasma in 10 of 12 animals and induced a variable decrease in the level of cell-associated SIV DNA in peripheral blood (average changes of 0.9-, 1.1-, 1.5-, and 3.7-fold for CD4(+) transitional memory [TTM], TCM, effector memory [TEM], and TSCM cells, respectively). ART-treated SIV-infected SMs showed (i) increased percentages of circulating CD4(+) TCM cells, (ii) increased levels of CD4(+) T cells in the rectal mucosa, and (iii) significant declines in the frequencies of HLA-DR(+) CD8(+) T cells in the blood and rectal mucosa. In addition, we observed that ART interruption resulted in rapid viral rebound in all SIV-infected SMs, indicating that the virus reservoir persists for at least a year under ART despite lower infection levels of CD4(+) TCM and TSCM cells than those seen in pathogenic SIV infections of macaques. Overall, these data indicate that ART induces specific immunological changes in SIV-infected SMs, thus suggesting that virus replication affects immune function even in the context of this clinically benign infection. IMPORTANCE: Studies of natural, nonpathogenic simian immunodeficiency virus (SIV) infection of African monkeys have provided important insights into the mechanisms responsible for the progression to AIDS during pathogenic human immunodeficiency virus (HIV) infection of humans and SIV infection of Asian macaques. In this study, for the first time, we treated SIV-infected sooty mangabeys, a natural host for the infection, with a potent antiretroviral therapy (ART) regimen for periods ranging from 2 to 12 months and monitored in detail how suppression of virus replication affected the main virological and immunological features of this nonpathogenic infection. The observed findings provide novel information on both the pathogenesis of residual immunological disease under ART during pathogenic infection and the mechanisms involved in virus persistence during primate lentiviral infections.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Cercocebus atys , Lentivirus Infections/veterinary , Primate Diseases/drug therapy , Simian Immunodeficiency Virus/drug effects , Animals , Blood/immunology , Blood/virology , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lentivirus Infections/drug therapy , Lentivirus Infections/pathology , Lentivirus Infections/virology , Primate Diseases/pathology , Primate Diseases/virology , Simian Immunodeficiency Virus/isolation & purification , Viral Load
13.
Nature ; 531(7595): 523-527, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26982722

ABSTRACT

The integrated stress response (ISR) is a homeostatic mechanism by which eukaryotic cells sense and respond to stress-inducing signals, such as amino acid starvation. General controlled non-repressed (GCN2) kinase is a key orchestrator of the ISR, and modulates protein synthesis in response to amino acid starvation. Here we demonstrate in mice that GCN2 controls intestinal inflammation by suppressing inflammasome activation. Enhanced activation of ISR was observed in intestinal antigen presenting cells (APCs) and epithelial cells during amino acid starvation, or intestinal inflammation. Genetic deletion of Gcn2 (also known as Eif2ka4) in CD11c(+) APCs or intestinal epithelial cells resulted in enhanced intestinal inflammation and T helper 17 cell (TH17) responses, owing to enhanced inflammasome activation and interleukin (IL)-1ß production. This was caused by reduced autophagy in Gcn2(-/-) intestinal APCs and epithelial cells, leading to increased reactive oxygen species (ROS), a potent activator of inflammasomes. Thus, conditional ablation of Atg5 or Atg7 in intestinal APCs resulted in enhanced ROS and TH17 responses. Furthermore, in vivo blockade of ROS and IL-1ß resulted in inhibition of TH17 responses and reduced inflammation in Gcn2(-/-) mice. Importantly, acute amino acid starvation suppressed intestinal inflammation via a mechanism dependent on GCN2. These results reveal a mechanism that couples amino acid sensing with control of intestinal inflammation via GCN2.


Subject(s)
Amino Acids/metabolism , Colitis/metabolism , Inflammasomes/antagonists & inhibitors , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestines/pathology , Protein Serine-Threonine Kinases/metabolism , Amino Acids/administration & dosage , Amino Acids/deficiency , Amino Acids/pharmacology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Autophagy , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Colitis/etiology , Colitis/pathology , Colitis/prevention & control , Disease Models, Animal , Epithelial Cells/metabolism , Female , Humans , Inflammasomes/metabolism , Inflammation/etiology , Inflammation/pathology , Inflammation/prevention & control , Interleukin-1beta/immunology , Male , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Stress, Physiological , Th17 Cells/immunology , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/metabolism
14.
J Virol ; 89(17): 8677-86, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26063417

ABSTRACT

UNLABELLED: Numerous studies have demonstrated that CD8(+) T lymphocytes suppress virus replication during human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) infection. However, the mechanisms underlying this activity of T cells remain incompletely understood. Here, we conducted CD8(+) T lymphocyte depletion in 15 rhesus macaques (RMs) infected intravenously (i.v.) with SIVmac239. At day 70 postinfection, the animals (10 progressors with high viremia and 5 controllers with low viremia) were CD8 depleted by i.v. administration of the antibody M-T807R1. As expected, CD8 depletion resulted in increased virus replication, more prominently in controllers than progressors, which correlated inversely with predepletion viremia. Of note, the feature of CD8(+) T lymphocyte predepletion that correlated best with the increase in viremia postdepletion was the level of CD8(+) T-bet(+) lymphocytes. We next found that CD8 depletion resulted in a homogenous increase of SIV RNA in superficial and mesenteric lymph nodes, spleen, and the gastrointestinal tract of both controllers and progressors. Interestingly, the level of SIV DNA increased postdepletion in both CD4(+) central memory T lymphocytes (TCM) and CD4(+) effector memory T lymphocytes (TEM) in progressor RMs but decreased in the CD4(+) TCM of 4 out of 5 controllers. Finally, we found that CD8 depletion is associated with a greater increase in CD4(+) T lymphocyte activation (measured by Ki-67 expression) in controllers than in progressors. Overall, these data reveal a differential impact of CD8(+) T lymphocyte depletion between controller and progressor SIV-infected RMs, emphasizing the complexity of the in vivo antiviral role of CD8(+) T lymphocytes. IMPORTANCE: In this study, we further dissect the impact of CD8(+) T lymphocytes on HIV/SIV replication during SIV infection. CD8(+) T lymphocyte depletion leads to a relatively homogenous increase in viral replication in peripheral blood and tissues. CD8(+) T lymphocyte depletion resulted in a more prominent increase in viral loads and CD4(+) T lymphocyte activation in controllers than in progressors. Interestingly, we found T-bet expression on CD8(+) T lymphocytes to be the best predictor of viral load increase following depletion. The levels of SIV DNA increase postdepletion in both CD4(+) TCM and TEM in progressor RMs but decrease in the CD4(+) TCM of controllers. The findings described in this study provide key insights into the differential functions of CD8(+) T lymphocytes in controller and progressor RMs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Lymphocyte Depletion , Simian Immunodeficiency Virus/immunology , Animals , DNA, Viral/genetics , Female , Gastrointestinal Tract/virology , Immunologic Memory/immunology , Lymph Nodes/virology , Macaca mulatta , RNA, Viral/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Spleen/virology , Viral Load , Viremia/veterinary , Viremia/virology , Virus Replication/immunology
15.
Proc Natl Acad Sci U S A ; 112(12): E1480-9, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25730868

ABSTRACT

HIV-1 infection is characterized by varying degrees of chronic immune activation and disruption of T-cell homeostasis, which impact the rate of disease progression. A deeper understanding of the factors that influence HIV-1-induced immunopathology and subsequent CD4(+) T-cell decline is critical to strategies aimed at controlling or eliminating the virus. In an analysis of 127 acutely infected Zambians, we demonstrate a dramatic and early impact of viral replicative capacity (vRC) on HIV-1 immunopathogenesis that is independent of viral load (VL). Individuals infected with high-RC viruses exhibit a distinct inflammatory cytokine profile as well as significantly elevated T-cell activation, proliferation, and CD8(+) T-cell exhaustion, during the earliest months of infection. Moreover, the vRC of the transmitted virus is positively correlated with the magnitude of viral burden in naive and central memory CD4(+) T-cell populations, raising the possibility that transmitted viral phenotypes may influence the size of the initial latent viral reservoir. Taken together, these findings support an unprecedented role for the replicative fitness of the founder virus, independent of host protective genes and VL, in influencing multiple facets of HIV-1-related immunopathology, and that a greater focus on this parameter could provide novel approaches to clinical interventions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/physiology , Virus Replication , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , Cohort Studies , Cytokines/blood , Cytokines/metabolism , Disease Progression , Female , HIV Infections/blood , Homeostasis , Humans , Immunologic Memory , Inflammation , Kaplan-Meier Estimate , Lymphocyte Activation , Male , Molecular Sequence Data , Time Factors , Viral Load
16.
Proc Natl Acad Sci U S A ; 112(10): 3050-5, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25713354

ABSTRACT

CD8 T cells are a potent tool for eliminating intracellular pathogens and tumor cells. Thus, eliciting robust CD8 T-cell immunity is the basis for many vaccines under development. However, the relationship between antigen load and the magnitude of the CD8 T-cell response is not well-described in a human immune response. Here we address this issue by quantifying viral load and the CD8 T-cell response in a cohort of 80 individuals immunized with the live attenuated yellow fever vaccine (YFV-17D) by sampling peripheral blood at days 0, 1, 2, 3, 5, 7, 9, 11, 14, 30, and 90. When the virus load was below a threshold (peak virus load < 225 genomes per mL, or integrated virus load < 400 genome days per mL), the magnitude of the CD8 T-cell response correlated strongly with the virus load (R(2) ∼ 0.63). As the virus load increased above this threshold, the magnitude of the CD8 T-cell responses saturated. Recent advances in CD8 T-cell-based vaccines have focused on replication-incompetent or single-cycle vectors. However, these approaches deliver relatively limited amounts of antigen after immunization. Our results highlight the requirement that T-cell-based vaccines should deliver sufficient antigen during the initial period of the immune response to elicit a large number of CD8 T cells that may be needed for protection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Viral Load , Yellow Fever Vaccine/immunology , Cohort Studies , Gene Expression Profiling , Humans , Yellow fever virus/genetics , Yellow fever virus/immunology , Yellow fever virus/isolation & purification
17.
PLoS Pathog ; 10(9): e1004406, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25254512

ABSTRACT

Despite many advances in AIDS research, a cure for HIV infection remains elusive. Here, we performed autologous hematopoietic stem cell transplantation (HSCT) in three Simian/Human Immunodeficiency Virus (SHIV)-infected, antiretroviral therapy (ART)-treated rhesus macaques (RMs) using HSCs collected prior to infection and compared them to three SHIV-infected, ART-treated, untransplanted control animals to assess the effect of conditioning and autologous HSCT on viral persistence. As expected, ART drastically reduced virus replication, below 100 SHIV-RNA copies per ml of plasma in all animals. After several weeks on ART, experimental RMs received myeloablative total body irradiation (1080 cGy), which resulted in the depletion of 94-99% of circulating CD4+ T-cells, and low to undetectable SHIV-DNA levels in peripheral blood mononuclear cells. Following HSC infusion and successful engraftment, ART was interrupted (40-75 days post-transplant). Despite the observed dramatic reduction of the peripheral blood viral reservoir, rapid rebound of plasma viremia was observed in two out of three transplanted RMs. In the third transplanted animal, plasma SHIV-RNA and SHIV DNA in bulk PBMCs remained undetectable at week two post-ART interruption. No further time-points could be assessed as this animal was euthanized for clinical reasons; however, SHIV-DNA could be detected in this animal at necropsy in sorted circulating CD4+ T-cells, spleen and lymph nodes but not in the gastro-intestinal tract or tonsils. Furthermore, SIV DNA levels post-ART interruption were equivalent in several tissues in transplanted and control animals. While persistence of virus reservoir was observed despite myeloablation and HSCT in the setting of short term ART, this experiment demonstrates that autologous HSCT can be successfully performed in SIV-infected ART-treated RMs offering a new experimental in vivo platform to test innovative interventions aimed at curing HIV infection in humans.


Subject(s)
Anti-Retroviral Agents/pharmacology , HIV Reverse Transcriptase/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Leukocytes, Mononuclear/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Virus Latency/drug effects , Animals , Flow Cytometry , Immunophenotyping , Leukocytes, Mononuclear/drug effects , Macaca mulatta , Male , RNA, Viral/genetics , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/genetics , Transplantation, Autologous , Virus Replication
18.
J Immunol ; 193(9): 4527-36, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25246494

ABSTRACT

The inhibitory receptor programmed death-1 (PD-1) has been shown to regulate CD8 T cell function during chronic SIV infection; however, its role on CD4 T cells, specifically in the gut-associated lymphoid tissue, is less well understood. In this study, we show that a subset of CD4 T cells expresses high levels of PD-1 (PD-1(hi)) in the rectal mucosa, a preferential site of virus replication. The majority of these PD-1(hi) CD4 T cells expressed Bcl-6 and CXCR5, markers characteristic of T follicular helper cells in the lymph nodes. Following a pathogenic SIV infection, the frequency of PD-1(hi) cells (as a percentage of CD4 T cells) dramatically increased in the rectal mucosa; however, a significant fraction of them did not express CXCR5. Furthermore, only a small fraction of PD-1(hi) cells expressed CCR5, and despite this low level of viral coreceptor expression, a significant fraction of these cells were productively infected. Interestingly, vaccinated SIV controllers did not present with this aberrant PD-1(hi) CD4 T cell enrichment, and this lack of enrichment was associated with the presence of higher frequencies of SIV-specific granzyme B(+) CD8 T cells within the lymphoid tissue, suggesting a role for antiviral CD8 T cells in limiting aberrant expansion of PD-1(hi) CD4 T cells. These results highlight the importance of developing vaccines that enhance antiviral CD8 T cells at sites of preferential viral replication and support the need for developing therapeutic interventions that limit expansion of SIV(+)PD-1(hi) CD4 T cells at mucosal sites as a means to enhance viral control.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Peyer's Patches/immunology , Peyer's Patches/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Animals , Antigens, Surface/metabolism , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Survival , Gene Expression , Immunophenotyping , Interleukin-2/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/virology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macaca mulatta , Peyer's Patches/metabolism , Phenotype , Programmed Cell Death 1 Receptor/metabolism , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Rectum/immunology , Rectum/metabolism , Rectum/virology , Simian Acquired Immunodeficiency Syndrome/metabolism , Viral Load , Virus Replication
19.
J Clin Invest ; 124(7): 3147-58, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24911151

ABSTRACT

BACKGROUND: Defining the parameters that modulate vaccine responses in African populations will be imperative to design effective vaccines for protection against HIV, malaria, tuberculosis, and dengue virus infections. This study aimed to evaluate the contribution of the patient-specific immune microenvironment to the response to the licensed yellow fever vaccine 17D (YF-17D) in an African cohort. METHODS: We compared responses to YF-17D in 50 volunteers in Entebbe, Uganda, and 50 volunteers in Lausanne, Switzerland. We measured the CD8+ T cell and B cell responses induced by YF-17D and correlated them with immune parameters analyzed by flow cytometry prior to vaccination. RESULTS: We showed that YF-17D-induced CD8+ T cell and B cell responses were substantially lower in immunized individuals from Entebbe compared with immunized individuals from Lausanne. The impaired vaccine response in the Entebbe cohort associated with reduced YF-17D replication. Prior to vaccination, we observed higher frequencies of exhausted and activated NK cells, differentiated T and B cell subsets and proinflammatory monocytes, suggesting an activated immune microenvironment in the Entebbe volunteers. Interestingly, activation of CD8+ T cells and B cells as well as proinflammatory monocytes at baseline negatively correlated with YF-17D-neutralizing antibody titers after vaccination. Additionally, memory T and B cell responses in preimmunized volunteers exhibited reduced persistence in the Entebbe cohort but were boosted by a second vaccination. CONCLUSION: Together, these results demonstrate that an activated immune microenvironment prior to vaccination impedes efficacy of the YF-17D vaccine in an African cohort and suggest that vaccine regimens may need to be boosted in African populations to achieve efficient immunity. TRIAL REGISTRATION: Registration is not required for observational studies. FUNDING: This study was funded by Canada's Global Health Research Initiative, Defense Threat Reduction Agency, National Institute of Allergy and Infectious Diseases, Bill & Melinda Gates Foundation, and United States Agency for International Development.


Subject(s)
Yellow Fever Vaccine/immunology , Yellow Fever/immunology , Yellow Fever/prevention & control , Adaptive Immunity , Adult , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cohort Studies , Female , Humans , Immunity, Cellular , Immunity, Humoral , Immunity, Innate , Immunization, Secondary , Lymphocyte Activation , Male , Middle Aged , Monocytes/immunology , Switzerland , Uganda , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Virus Replication/immunology , Yellow Fever/virology , Yellow Fever Vaccine/administration & dosage , Yellow fever virus/immunology , Yellow fever virus/physiology , Young Adult
20.
J Immunol ; 192(10): 4666-73, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24729621

ABSTRACT

Recent studies have identified a subset of memory T cells with stem cell-like properties (T(SCM)) that include increased longevity and proliferative potential. In this study, we examined the dynamics of CD4(+) T(SCM) during pathogenic SIV infection of rhesus macaques (RM) and nonpathogenic SIV infection of sooty mangabeys (SM). Whereas SIV-infected RM show selective numeric preservation of CD4(+) T(SCM), SIV infection induced a complex perturbation of these cells defined by depletion of CD4(+)CCR5(+) T(SCM), increased rates of CD4(+) T(SCM) proliferation, and high levels of direct virus infection. The increased rates of CD4(+) T(SCM) proliferation in SIV-infected RM correlated inversely with the levels of central memory CD4(+) T cells. In contrast, nonpathogenic SIV infection of SM evidenced preservation of both CD4(+) T(SCM) and CD4(+) central memory T cells, with normal levels of CD4(+) T(SCM) proliferation, and lack of selective depletion of CD4(+)CCR5(+) T(SCM). Importantly, SIV DNA was below the detectable limit in CD4(+) T(SCM) from 8 of 10 SIV-infected SM. We propose that increased proliferation and infection of CD4(+) T(SCM) may contribute to the pathogenesis of SIV infection in RM.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Cercocebus atys , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...