Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
1.
JCI Insight ; 8(14)2023 07 24.
Article in English | MEDLINE | ID: mdl-37485874

ABSTRACT

Resolution of T cell activation and inflammation is a key determinant of the lack of SIV disease progression in African green monkeys (AGMs). Although frequently considered together, T cell activation occurs in response to viral stimulation of acquired immunity, while inflammation reflects innate immune responses to mucosal injury. We dissociated T cell activation from inflammation through regulatory T cell (Treg) depletion with Ontak (interleukin-2 coupled with diphtheria toxin) during early SIV infection of AGMs. This intervention abolished control of T cell immune activation beyond the transition from acute to chronic infection. Ontak had no effect on gut barrier integrity, microbial translocation, inflammation, and hypercoagulation, despite increasing T cell activation. Ontak administration increased macrophage counts yet decreased their activation. Persistent T cell activation influenced SIV pathogenesis, shifting the ramp-up in viral replication to earlier time points, prolonging the high levels of replication, and delaying CD4+ T cell restoration yet without any clinical or biological sign of disease progression in Treg-depleted AGMs. Thus, by inducing T cell activation without damaging mucosal barrier integrity, we showed that systemic T cell activation per se is not sufficient to drive disease progression, which suggests that control of systemic inflammation (likely through maintenance of gut integrity) is the key determinant of lack of disease progression in natural hosts of SIVs.


Subject(s)
Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Chlorocebus aethiops , Disease Progression , CD4-Positive T-Lymphocytes , Inflammation
2.
PLoS Pathog ; 18(6): e1010507, 2022 06.
Article in English | MEDLINE | ID: mdl-35714165

ABSTRACT

The HIV/SIV envelope glycoprotein (Env) cytoplasmic domain contains a highly conserved Tyr-based trafficking signal that mediates both clathrin-dependent endocytosis and polarized sorting. Despite extensive analysis, the role of these functions in viral infection and pathogenesis is unclear. An SIV molecular clone (SIVmac239) in which this signal is inactivated by deletion of Gly-720 and Tyr-721 (SIVmac239ΔGY), replicates acutely to high levels in pigtail macaques (PTM) but is rapidly controlled. However, we previously reported that rhesus macaques and PTM can progress to AIDS following SIVmac239ΔGY infection in association with novel amino acid changes in the Env cytoplasmic domain. These included an R722G flanking the ΔGY deletion and a nine nucleotide deletion encoding amino acids 734-736 (ΔQTH) that overlaps the rev and tat open reading frames. We show that molecular clones containing these mutations reconstitute signals for both endocytosis and polarized sorting. In one PTM, a novel genotype was selected that generated a new signal for polarized sorting but not endocytosis. This genotype, together with the ΔGY mutation, was conserved in association with high viral loads for several months when introduced into naïve PTMs. For the first time, our findings reveal strong selection pressure for Env endocytosis and particularly for polarized sorting during pathogenic SIV infection in vivo.


Subject(s)
Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Endocytosis , Gene Products, env/genetics , Macaca mulatta/metabolism , Macaca nemestrina , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism
3.
Sci Rep ; 9(1): 12802, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31488856

ABSTRACT

Recent data in a nonhuman primate model showed that infants postnatally infected with Zika virus (ZIKV) were acutely susceptible to high viremia and neurological damage, suggesting the window of vulnerability extends beyond gestation. In this pilot study, we addressed the susceptibility of two infant rhesus macaques born healthy to dams infected with Zika virus during pregnancy. Passively acquired neutralizing antibody titers dropped below detection limits between 2 and 3 months of age, while binding antibodies remained detectable until viral infection at 5 months. Acute serum viremia was comparatively lower than adults infected with the same Brazilian isolate of ZIKV (n = 11 pregnant females, 4 males, and 4 non-pregnant females). Virus was never detected in cerebrospinal fluid nor in neural tissues at necropsy two weeks after infection. However, viral RNA was detected in lymph nodes, confirming some tissue dissemination. Though protection was not absolute and our study lacks an important comparison with postnatally infected infants born to naïve dams, our data suggest infants born healthy to infected mothers may harbor a modest but important level of protection from postnatally acquired ZIKV for several months after birth, an encouraging result given the potentially severe infection outcomes of this population.


Subject(s)
Infectious Disease Transmission, Vertical , Macaca mulatta , Pregnancy Complications, Infectious/veterinary , Zika Virus Infection/transmission , Animals , Animals, Newborn/immunology , Animals, Newborn/virology , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Female , Male , Pilot Projects , Pregnancy , Pregnancy Complications, Infectious/virology , Zika Virus , Zika Virus Infection/immunology , Zika Virus Infection/virology
4.
JCI Insight ; 4(4)2019 02 21.
Article in English | MEDLINE | ID: mdl-30830861

ABSTRACT

Newly emerging viruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV), Middle Eastern respiratory syndrome CoVs (MERS-CoV), and H7N9, cause fatal acute lung injury (ALI) by driving hypercytokinemia and aggressive inflammation through mechanisms that remain elusive. In SARS-CoV/macaque models, we determined that anti-spike IgG (S-IgG), in productively infected lungs, causes severe ALI by skewing inflammation-resolving response. Alveolar macrophages underwent functional polarization in acutely infected macaques, demonstrating simultaneously both proinflammatory and wound-healing characteristics. The presence of S-IgG prior to viral clearance, however, abrogated wound-healing responses and promoted MCP1 and IL-8 production and proinflammatory monocyte/macrophage recruitment and accumulation. Critically, patients who eventually died of SARS (hereafter referred to as deceased patients) displayed similarly accumulated pulmonary proinflammatory, absence of wound-healing macrophages, and faster neutralizing antibody responses. Their sera enhanced SARS-CoV-induced MCP1 and IL-8 production by human monocyte-derived wound-healing macrophages, whereas blockade of FcγR reduced such effects. Our findings reveal a mechanism responsible for virus-mediated ALI, define a pathological consequence of viral specific antibody response, and provide a potential target for treatment of SARS-CoV or other virus-mediated lung injury.


Subject(s)
Acute Lung Injury/immunology , Antibodies, Viral/immunology , Immunoglobulin G/immunology , Severe Acute Respiratory Syndrome/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Spike Glycoprotein, Coronavirus/immunology , Acute Lung Injury/blood , Acute Lung Injury/virology , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/administration & dosage , Antibodies, Viral/blood , Cell Line , Disease Models, Animal , Female , Humans , Immunization, Passive/methods , Immunoglobulin G/administration & dosage , Immunoglobulin G/blood , Lung/immunology , Lung/pathology , Lung/virology , Macaca mulatta , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Middle Aged , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Severe Acute Respiratory Syndrome/blood , Severe Acute Respiratory Syndrome/virology , Spike Glycoprotein, Coronavirus/genetics , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Young Adult
5.
Nat Commun ; 9(1): 1624, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29691387

ABSTRACT

Zika virus (ZIKV) infection of pregnant women is associated with pathologic complications of fetal development. Here, we infect pregnant rhesus macaques (Macaca mulatta) with a minimally passaged ZIKV isolate from Rio de Janeiro, where a high rate of fetal development complications was observed. The infection of pregnant macaques with this virus results in maternal viremia, virus crossing into the amniotic fluid (AF), and in utero fetal deaths. We also treated three additional ZIKV-infected pregnant macaques with a cocktail of ZIKV-neutralizing human monoclonal antibodies (nmAbs) at peak viremia. While the nmAbs can be effective in clearing the virus from the maternal sera of treated monkeys, it is not sufficient to clear ZIKV from AF. Our report suggests that ZIKV from Brazil causes fetal demise in non-human primates (NHPs) without additional mutations or confounding co-factors. Treatment with a neutralizing anti-ZIKV nmAb cocktail is insufficient to fully stop vertical transmission.


Subject(s)
Antibodies, Viral/administration & dosage , Pregnancy Complications/drug therapy , Zika Virus Infection/drug therapy , Zika Virus/physiology , Animals , Antibodies, Neutralizing/administration & dosage , Female , Fetal Death , Humans , Macaca mulatta , Pregnancy , Pregnancy Complications/mortality , Pregnancy Complications/virology , Zika Virus/drug effects , Zika Virus/genetics , Zika Virus Infection/mortality , Zika Virus Infection/virology
6.
Proc Natl Acad Sci U S A ; 115(1): E62-E71, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29255022

ABSTRACT

Mycobacterium tuberculosis continues to cause devastating levels of mortality due to tuberculosis (TB). The failure to control TB stems from an incomplete understanding of the highly specialized strategies that M. tuberculosis utilizes to modulate host immunity and thereby persist in host lungs. Here, we show that M. tuberculosis induced the expression of indoleamine 2,3-dioxygenase (IDO), an enzyme involved in tryptophan catabolism, in macrophages and in the lungs of animals (mice and macaque) with active disease. In a macaque model of inhalation TB, suppression of IDO activity reduced bacterial burden, pathology, and clinical signs of TB disease, leading to increased host survival. This increased protection was accompanied by increased lung T cell proliferation, induction of inducible bronchus-associated lymphoid tissue and correlates of bacterial killing, reduced checkpoint signaling, and the relocation of effector T cells to the center of the granulomata. The enhanced killing of M. tuberculosis in macrophages in vivo by CD4+ T cells was also replicated in vitro, in cocultures of macaque macrophages and CD4+ T cells. Collectively, these results suggest that there exists a potential for using IDO inhibition as an effective and clinically relevant host-directed therapy for TB.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Lung/immunology , Mycobacterium tuberculosis/immunology , Tryptophan/immunology , Tuberculoma/immunology , Tuberculosis, Pulmonary/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Granuloma/immunology , Granuloma/pathology , Lung/pathology , Macaca mulatta , Macrophages/immunology , Macrophages/pathology , Mycobacterium tuberculosis/pathogenicity , Tuberculoma/pathology , Tuberculosis, Pulmonary/pathology
7.
Curr HIV Res ; 16(1): 29-40, 2018.
Article in English | MEDLINE | ID: mdl-29173176

ABSTRACT

BACKGROUND: Cellular tropism of human immunodeficiency virus (HIV-1) is closely linked to interactions between the viral envelope glycoprotein (Env) with CD4 and chemokine receptor family members, CCR5 and CXCR4. This interaction plays a key role in determining anatomic sites that are infected in vivo and the cascade of early and late events that result in chronic immune activation, immunosuppression and ultimately, AIDS. CD4+ T cells are critical to adaptive immune responses, and their early and rapid infection in gut lamina propria and secondary lymphoid tissues in susceptible hosts likely contributes to viral persistence and progression to disease. CD4+ macrophages are also infected, although their role in HIV-1 pathogenesis is more controversial. METHODS: Pathogenic infection by simian immunodeficiency viruses (SIV) in Asian macaques as models of HIV-1 infection has enabled the impact of cellular tropism on pathogenesis to be directly probed. This review will highlight examples in which experimental interventions during SIV infection or the introduction of viral mutations have altered cellular tropism and, subsequently, pathogenesis. RESULTS: Alterations to the interaction of Env and its cellular receptors has been shown to result in changes to CD4 dependence, coreceptor specificity, and viral tropism for gut CD4+ T cells and macrophages. CONCLUSION: Collectively, these findings have yielded novel insights into the critical role of the viral Env and tropism as a driver of pathogenesis and host control and have helped to identify new areas for targeted interventions in therapy and prevention of HIV-1 infection.


Subject(s)
Disease Susceptibility , Gene Products, env/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Viral Tropism , Animals , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Receptors, CCR5/metabolism
8.
Ann N Y Acad Sci ; 1408(1): 79-89, 2017 11.
Article in English | MEDLINE | ID: mdl-29239481

ABSTRACT

A reduction in the burden of schistosomiasis is potentially achievable by integrating a schistosomiasis vaccine with current control measures. Here, we determine parasite-specific in vitro responses of B, T, and NK cells from naive uninfected rhesus macaques to Schistosoma mansoni (Sm) egg (SmEA) and worm antigen (SmWA) preparations isolated from infected baboons. Pronounced B cell responses to SmEA and NK cell responses to both SmEA and SmWA were observed. High levels of IL-2 and IL-21 responses against Sm antigens were observed in T and non-T cells of lymph nodes (LNs) and gut lamina propria-derived lymphocytes (LPLs). Data analysis showed multifunctionality of LN-derived CD4+ , CD8+ , and CD4+ CD8+ double positive T cells against either SmWA or SmWA+SmEA antigen preparations. Distinct SmEA-specific multifunctional responses were observed in gut LPLs, suggesting simultaneous responses against egg antigens. These data provide insight into the immune effectors involved in schistosome responses by rhesus macaques.


Subject(s)
Antigens, Helminth/immunology , Monkey Diseases/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/veterinary , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukins/immunology , Interleukins/metabolism , Killer Cells, Natural/immunology , Lymph Nodes/immunology , Macaca mulatta , Monkey Diseases/parasitology , Papio , Remission, Spontaneous , Schistosoma mansoni/physiology , Schistosomiasis mansoni/parasitology
9.
ILAR J ; 58(2): 160-171, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29228218

ABSTRACT

Research using nonhuman primates (NHPs) as models for human immunodeficiency virus (HIV) infection and acquired immunodeficiency syndrome (AIDS) has resulted in tremendous achievements not only in the prevention and treatment of HIV, but also in biomedical research more broadly. Once considered a death sentence, HIV infection is now fairly well controlled with combination antiretroviral treatments, almost all of which were first tested for efficacy and safety in nonhuman primates or other laboratory animals. Research in NHP has led to "dogma changing" discoveries in immunology, infectious disease, and even our own genetics. We now know that many of our genes are retroviral remnants, or developed in response to archaic HIV-like retroviral infections. Early studies involving blood from HIV patients and in experiments in cultured tissues contributed to confusion regarding the cause of AIDS and impeded progress in the development of effective interventions. Research on the many retroviruses of different NHP species have broadened our understanding of human immunology and perhaps even our origins and evolution as a species. In combination with recent advances in molecular biology and computational analytics, research in NHPs has unique potential for discoveries that will directly lead to new cures for old human and animal diseases, including HIV/AIDS.


Subject(s)
Acquired Immunodeficiency Syndrome/pathology , HIV Infections/pathology , Animals , Coinfection , Disease Models, Animal , Humans , Macaca fascicularis , Macaca mulatta , Mycobacterium tuberculosis/pathogenicity , Primates , Tuberculosis/microbiology , Tuberculosis/pathology
10.
Am J Pathol ; 187(12): 2811-2820, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28935575

ABSTRACT

Failure to replace Bacille Calmette-Guerin vaccines with efficacious anti-tuberculosis (TB) vaccines have prompted outside-the-box thinking, including pulmonary vaccination to elicit local immunity. Inhalational MtbΔsigH, a stress-response-attenuated strain, protected against lethal TB in macaques. While live mycobacterial vaccines show promising efficacy, HIV co-infection and the resulting immunodeficiency prompts safety concerns about their use. We assessed the persistence and safety of MtbΔsigH, delivered directly to the lungs, in the setting of HIV co-infection. Macaques were aerosol-vaccinated with ΔsigH and subsequently challenged with SIVmac239. Bronchoalveolar lavage and tissues were sampled for mycobacterial persistence, pathology, and immune correlates. Only 35% and 3.5% of lung samples were positive for live bacilli and granulomas, respectively. Our results therefore suggest that the nonpathologic infection of macaque lungs by ΔsigH was not reactivated by simian immunodeficiency virus, despite high viral levels and massive ablation of pulmonary CD4+ T cells. Protective pulmonary responses were retained, including vaccine-induced bronchus-associated lymphoid tissue and CD8+ effector memory T cells. Despite acute simian immunodeficiency virus infection, all animals remained asymptomatic of pulmonary TB. These findings highlight the efficacy of mucosal vaccination via this attenuated strain and will guide its further development to potentially combat TB in HIV-endemic areas. Our results also suggest that a lack of pulmonary pathology is a key correlate of the safety of live mycobacterial vaccines.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/complications , Tuberculosis Vaccines/pharmacology , Tuberculosis/prevention & control , Virus Activation/drug effects , Administration, Inhalation , Animals , Coinfection , HIV , Macaca mulatta , Mycobacterium tuberculosis , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/physiology , Tuberculosis/complications , Vaccines, Attenuated/pharmacology
11.
Mediators Inflamm ; 2017: 5186904, 2017.
Article in English | MEDLINE | ID: mdl-28839349

ABSTRACT

Profound loss of CD4+ T cells, progressive impairment of the immune system, inflammation, and sustained immune activation are the characteristics of human immunodeficiency virus-1 (HIV-1) infection. Innate immune responses respond immediately from the day of HIV infection, and a thorough understanding of the interaction between several innate immune cells and HIV-1 is essential to determine to what extent those cells play a crucial role in controlling HIV-1 in vivo. Defensins, divided into the three subfamilies α-, ß-, and θ-defensins based on structure and disulfide linkages, comprise a critical component of the innate immune response and exhibit anti-HIV-1 activities and immunomodulatory capabilities. In humans, only α- and ß-defensins are expressed in various tissues and have broad impacts on HIV-1 transmission, replication, and disease progression. θ-defensins have been identified as functional peptides in Old World monkeys, but not in humans. Instead, θ-defensins exist only as pseudogenes in humans, chimpanzees, and gorillas. The use of the synthetic θ-defensin peptide "retrocyclin" as an antiviral therapy was shown to be promising, and further research into the development of defensin-based HIV-1 therapeutics is needed. This review focuses on the role of defensins in HIV-1 pathogenesis and highlights future research efforts that warrant investigation.


Subject(s)
HIV Infections/metabolism , HIV Infections/pathology , Animals , Biomarkers/metabolism , C-Reactive Protein/metabolism , Defensins/metabolism , Female , Fibrinogen/metabolism , Humans , Male
12.
ILAR J ; 58(2): 151-159, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28575319

ABSTRACT

Infection with Mycobacterium tuberculosis predominantly establishes subclinical latent infection over the lifetime of an individual, with a fraction of infected individuals rapidly progressing to active disease. The immune control in latent infection can be perturbed by comorbidities such as diabetes mellitus, obesity, smoking, and coinfection with helminthes or HIV. Modeling the varying aspects of natural infection remains incomplete when using zebrafish and mice. However, the nonhuman primate model of tuberculosis offers a unique and accurate model to investigate host responses to infection, test novel therapeutics, and thoroughly assess preclinical vaccine candidates. Rhesus macaques and cynomolgus macaques manifest the full gamut of clinical and pathological findings in human Mycobacterium tuberculosis infection, including the ability to co-infect macaques with Simian Immunodeficiency Virus to model HIV co-infection. Here we discuss advanced techniques to assay various clinical outcomes of the natural progression of infection as well as therapeutics in development and novel preclinical vaccines. Finally, we survey the translational aspects of nonhuman primate research and argue the urgent need to thoroughly examine preclinical therapeutics and vaccines using this model prior to clinical implementation.


Subject(s)
Translational Research, Biomedical/methods , Animals , Coinfection , HIV Infections/immunology , Macaca mulatta , Mice , Mycobacterium tuberculosis/pathogenicity , Simian Immunodeficiency Virus/pathogenicity , Tuberculosis/immunology , Tuberculosis/microbiology , Zebrafish
13.
JCI Insight ; 2(6): e91020, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28352660

ABSTRACT

Infection is the most common cause of mortality in early life, and immunization is the most promising biomedical intervention to reduce this burden. However, newborns fail to respond optimally to most vaccines. Adjuvantation is a key approach to enhancing vaccine immunogenicity, but responses of human newborn leukocytes to most candidate adjuvants, including most TLR agonists, are functionally distinct. Herein, we demonstrate that 3M-052 is a locally acting lipidated imidazoquinoline TLR7/8 agonist adjuvant in mice, which, when properly formulated, can induce robust Th1 cytokine production by human newborn leukocytes in vitro, both alone and in synergy with the alum-adjuvanted pneumococcal conjugate vaccine 13 (PCV13). When admixed with PCV13 and administered i.m. on the first day of life to rhesus macaques, 3M-052 dramatically enhanced generation of Th1 CRM-197-specific neonatal CD4+ cells, activation of newborn and infant Streptococcus pneumoniae polysaccharide-specific (PnPS-specific) B cells as well as serotype-specific antibody titers, and opsonophagocytic killing. Remarkably, a single dose at birth of PCV13 plus 0.1 mg/kg 3M-052 induced PnPS-specific IgG responses that were approximately 10-100 times greater than a single birth dose of PCV13 alone, rapidly exceeding the serologic correlate of protection, as early as 28 days of life. This potent immunization strategy, potentially effective with one birth dose, could represent a new paradigm in early life vaccine development.


Subject(s)
Pneumococcal Vaccines/administration & dosage , Pneumococcal Vaccines/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology , Adjuvants, Immunologic/pharmacology , Adult , Animals , B-Lymphocytes/immunology , Cells, Cultured , Humans , Infant, Newborn , Macaca mulatta , T-Lymphocytes/immunology
14.
Am J Respir Cell Mol Biol ; 56(5): 637-647, 2017 05.
Article in English | MEDLINE | ID: mdl-28135421

ABSTRACT

Although it is accepted that the environment within the granuloma profoundly affects Mycobacterium tuberculosis (Mtb) and infection outcome, our ability to understand Mtb gene expression in these niches has been limited. We determined intragranulomatous gene expression in human-like lung lesions derived from nonhuman primates with both active tuberculosis (ATB) and latent TB infection (LTBI). We employed a non-laser-based approach to microdissect individual lung lesions and interrogate the global transcriptome of Mtb within granulomas. Mtb genes expressed in classical granulomas with central, caseous necrosis, as well as within the caseum itself, were identified and compared with other Mtb lesions in animals with ATB (n = 7) or LTBI (n = 7). Results were validated using both an oligonucleotide approach and RT-PCR on macaque samples and by using human TB samples. We detected approximately 2,900 and 1,850 statistically significant genes in ATB and LTBI lesions, respectively (linear models for microarray analysis, Bonferroni corrected, P < 0.05). Of these genes, the expression of approximately 1,300 (ATB) and 900 (LTBI) was positively induced. We identified the induction of key regulons and compared our results to genes previously determined to be required for Mtb growth. Our results indicate pathways that Mtb uses to ensure its survival in a highly stressful environment in vivo. A large number of genes is commonly expressed in granulomas with ATB and LTBI. In addition, the enhanced expression of the dormancy survival regulon was a key feature of lesions in animals with LTBI, stressing its importance in the persistence of Mtb during the chronic phase of infection.


Subject(s)
Gene Expression Regulation, Bacterial , Genes, Bacterial , Granuloma/microbiology , Microbial Viability/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/physiology , Anaerobiosis , Animals , Gene Expression Profiling , Granuloma/pathology , Lung/microbiology , Lung/pathology , Macaca , Real-Time Polymerase Chain Reaction , Regulon/genetics , Reproducibility of Results , Transcriptome/genetics , Tuberculosis/genetics , Tuberculosis/microbiology , Tuberculosis/pathology
15.
Proc Natl Acad Sci U S A ; 113(38): E5636-44, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27601645

ABSTRACT

The synergy between Mycobacterium tuberculosis (Mtb) and HIV in coinfected patients has profoundly impacted global mortality because of tuberculosis (TB) and AIDS. HIV significantly increases rates of reactivation of latent TB infection (LTBI) to active disease, with the decline in CD4(+) T cells believed to be the major causality. In this study, nonhuman primates were coinfected with Mtb and simian immunodeficiency virus (SIV), recapitulating human coinfection. A majority of animals exhibited rapid reactivation of Mtb replication, progressing to disseminated TB and increased SIV-associated pathology. Although a severe loss of pulmonary CD4(+) T cells was observed in all coinfected macaques, a subpopulation of the animals was still able to prevent reactivation and maintain LTBI. Investigation of pulmonary immune responses and pathology in this cohort demonstrated that increased CD8(+) memory T-cell proliferation, higher granzyme B production, and expanded B-cell follicles correlated with protection from reactivation. Our findings reveal mechanisms that control SIV- and TB-associated pathology. These CD4-independent protective immune responses warrant further studies in HIV coinfected humans able to control their TB infection. Moreover, these findings will provide insight into natural immunity to Mtb and will guide development of novel vaccine strategies and immunotherapies.


Subject(s)
HIV Infections/immunology , Latent Tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Simian Immunodeficiency Virus/pathogenicity , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Cell Proliferation/genetics , Coinfection/virology , HIV/immunology , HIV/pathogenicity , HIV Infections/physiopathology , HIV Infections/virology , Humans , Immunologic Memory/genetics , Latent Tuberculosis/microbiology , Latent Tuberculosis/pathology , Latent Tuberculosis/virology , Lymphocyte Activation/immunology , Macaca mulatta/immunology , Macaca mulatta/microbiology , Macaca mulatta/virology , Mycobacterium tuberculosis/immunology , Simian Immunodeficiency Virus/immunology
16.
Curr Opin Virol ; 19: 92-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27544476

ABSTRACT

Non-human primate models of AIDS and neuroAIDS are the premiere model of HIV infection of the CNS and neuropathogenesis. This review discusses current SIV infection models of neuroAIDS emphasizing findings in the last two years. Consistent in these findings is the interplay between host factors that regulate immune responses to virus and viral replication. Several rapid models of AIDS with consistent CNS pathogenesis exist, each of which modulates by antibody treatment or viruses that cause rapid immune suppression and replicate well in macrophages. Consistent in all of these models are data underscoring the importance of monocyte and macrophage activation, infection and accumulation in the CNS.


Subject(s)
AIDS Dementia Complex , Central Nervous System/pathology , Central Nervous System/virology , Disease Models, Animal , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus/physiology , AIDS Dementia Complex/immunology , AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , Animals , Central Nervous System/immunology , Encephalitis, Viral/immunology , Encephalitis, Viral/pathology , Humans , Macaca mulatta , Macrophages/virology , Monocytes/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Virus Replication
17.
J Neurovirol ; 22(6): 831-839, 2016 12.
Article in English | MEDLINE | ID: mdl-27273075

ABSTRACT

Flaviviruses, including Zika and dengue (DENV), pose a serious global threat to human health. Of the 50+ million humans infected with DENV annually, approximately 1-3 % progress to severe disease manifestations, dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS). Several factors are suspected to mediate the course of infection and pathogenesis of DENV infection. DHF and DSS are associated with vascular leakage and neurological sequelae. Our hypothesis was that altered astrocyte activation and morphology would alter the dynamics of the extracellular space and hence, neuronal and vascular function. We investigated the mechanisms of neuropathogenesis DENV infection in rhesus macaques. There were decreased numbers of GFAP immunopositive astrocytes per unit area, although those that remained had increased arbor length and complexity. This was combined with structural hypertrophy of white matter astrocytes in the absence of increased vascular leakage. Combined, these studies show how even low-grade infection with DENV induces measurable changes within the parenchyma of infected individuals.


Subject(s)
Astrocytes/pathology , Dengue Virus/pathogenicity , Dengue/pathology , White Matter/pathology , Animals , Astrocytes/metabolism , Biomarkers/metabolism , Dengue/genetics , Dengue/metabolism , Dengue/virology , Dengue Virus/classification , Dengue Virus/genetics , Disease Models, Animal , Gene Expression , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Hypertrophy , Macaca mulatta , Serogroup , White Matter/metabolism , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
18.
J Transl Med ; 14(1): 148, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27230663

ABSTRACT

BACKGROUND: Many HIV infected individuals with suppressed viral loads experience chronic immune activation frequently developing neurological impairment designated as HIV associated neurocognitive disorder (HAND). Adjunctive therapies may reduce HIV associated inflammation and therefore decrease the occurrence of HAND. METHODS: We have conducted in vitro, animal and clinical studies of the neurokinin 1 receptor (NK1R) antagonist aprepitant in HIV/SIV infection. RESULTS: Aprepitant inhibits HIV infection of human macrophages ex vivo with an ED50 ~ 5 µM. When administered at 125 mg once daily for 12 months to SIV-infected rhesus macaques, aprepitant reduced viral load by approximately tenfold and produced anti-anxiolytic effects. The anti-viral and anti-anxiolytic effects occur at approximately the third month of dosing; and the effects are sustained throughout the duration of drug administration. Protein binding experiments in culture media and animal and human plasma indicate that the free fraction of aprepitant is lower than previously reported supporting usage of higher doses in vivo. The analysis of blood samples from HIV positive individuals treated for 2 weeks with aprepitant at doses up to 375 mg demonstrated reduced levels of pro-inflammatory cytokines including G-CSF, IL-6, IL-8 and TNFα. Decreased pro-inflammatory cytokines may reduce HIV comorbidities associated with chronic inflammation. CONCLUSIONS: Our results provide evidence for a unique combination of antiretroviral, anti-inflammatory and behavioral modulation properties of aprepitant in vitro and in vivo. These results provide robust support for a clinical exposure target above that recommended for chemotherapy-induced nausea and vomiting. Doses up to 375 mg once daily in HIV-infected patients still elicit sub-therapeutic exposure of aprepitant though effective plasma concentrations can be achievable by proper dose modulation.


Subject(s)
HIV Infections/drug therapy , Morpholines/therapeutic use , Neurokinin-1 Receptor Antagonists/therapeutic use , Receptors, Neurokinin-1/metabolism , Adolescent , Adult , Animals , Anti-Inflammatory Agents/pharmacology , Anxiety/complications , Aprepitant , Chemokines/metabolism , Female , HIV Infections/blood , HIV Infections/complications , Humans , Macaca mulatta , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Macrophages/virology , Male , Middle Aged , Monocytes/drug effects , Monocytes/pathology , Morpholines/blood , Neurokinin-1 Receptor Antagonists/pharmacology , Protein Binding/drug effects , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Substance P/pharmacology , Viral Load/drug effects , Virus Replication/drug effects , Young Adult
19.
J Virol ; 90(10): 4966-4980, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26937037

ABSTRACT

UNLABELLED: CD4 tropism is conserved among all primate lentiviruses and likely contributes to viral pathogenesis by targeting cells that are critical for adaptive antiviral immune responses. Although CD4-independent variants of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) have been described that can utilize the coreceptor CCR5 or CXCR4 in the absence of CD4, these viruses typically retain their CD4 binding sites and still can interact with CD4. We describe the derivation of a novel CD4-independent variant of pathogenic SIVmac239, termed iMac239, that was used to derive an infectious R5-tropic SIV lacking a CD4 binding site. Of the seven mutations that differentiate iMac239 from wild-type SIVmac239, a single change (D178G) in the V1/V2 region was sufficient to confer CD4 independence in cell-cell fusion assays, although other mutations were required for replication competence. Like other CD4-independent viruses, iMac239 was highly neutralization sensitive, although mutations were identified that could confer CD4-independent infection without increasing its neutralization sensitivity. Strikingly, iMac239 retained the ability to replicate in cell lines and primary cells even when its CD4 binding site had been ablated by deletion of a highly conserved aspartic acid at position 385, which, for HIV-1, plays a critical role in CD4 binding. iMac239, with and without the D385 deletion, exhibited an expanded host range in primary rhesus peripheral blood mononuclear cells that included CCR5(+) CD8(+) T cells. As the first non-CD4-tropic SIV, iMac239-ΔD385 will afford the opportunity to directly assess the in vivo role of CD4 targeting on pathogenesis and host immune responses. IMPORTANCE: CD4 tropism is an invariant feature of primate lentiviruses and likely plays a key role in pathogenesis by focusing viral infection onto cells that mediate adaptive immune responses and in protecting virions attached to cells from neutralizing antibodies. Although CD4-independent viruses are well described for HIV and SIV, these viruses characteristically retain their CD4 binding site and can engage CD4 if available. We derived a novel CD4-independent, CCR5-tropic variant of the pathogenic molecular clone SIVmac239, termed iMac239. The genetic determinants of iMac239's CD4 independence provide new insights into mechanisms that underlie this phenotype. This virus remained replication competent even after its CD4 binding site had been ablated by mutagenesis. As the first truly non-CD4-tropic SIV, lacking the capacity to interact with CD4, iMac239 will provide the unique opportunity to evaluate SIV pathogenesis and host immune responses in the absence of the immunomodulatory effects of CD4(+) T cell targeting and infection.


Subject(s)
CD4 Antigens/metabolism , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Viral Tropism , Virus Attachment , Animals , Antibodies, Neutralizing/immunology , Binding Sites , CD4 Antigens/immunology , CD8-Positive T-Lymphocytes/virology , Cell Line , Humans , Leukocytes, Mononuclear/virology , Macaca mulatta , Mutagenesis , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Viral Envelope Proteins/genetics , Virus Replication/genetics
20.
J Virol ; 90(3): 1578-87, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26608323

ABSTRACT

UNLABELLED: CD4(+) follicular T helper (Tfh) cells play a prominent role in humoral immune responses, but the mechanisms of their accumulation and infection in AIDS remain unclear. Here we found that germinal center (GC) Tfh cells, defined here as CXCR5(+) PD-1(HIGH) CD4(+) T cells, do not express the HIV coreceptor CCR5 yet serve as a latent reservoir in GCs. With disease progression, an expansion of GC Tfh cells is accompanied by increases in dysfunctional CD8(+) T cells. In contrast, Tfh precursor (CXCR5(-) CD4(+) T) cells in lymph nodes do express CCR5 and differentiate into GC Tfh cells following interleukin-6 (IL-6) and IL-21 stimulation, and viral DNA is detectable in fully differentiated GC Tfh cells ex vivo. This suggests that SIV-infected GC Tfh cells may be derived from Tfh precursor cell subsets that become infected in marginal zones and then migrate into GCs as fully mature GC Tfh cells that serve as persistent virus reservoirs. These findings suggest that viral persistence in lymph nodes drives compensatory differentiation, aberrant accumulation, and latent infection of GC Tfh cells, resulting in marked impairment of humoral immune responses. IMPORTANCE: Generation of antibodies that can effectively eliminate viruses requires interactions of B cells with highly specialized T cells in GCs of lymphoid tissues called follicular T helper cells. Here we show that in simian immunodeficiency virus infection, these cells are initially infected in a precursor stage that leads to alterations in their homing, accumulation, and function that may be responsible for the inability of human immunodeficiency virus-infected patients to generate effective antibody responses.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Germinal Center/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , T-Lymphocyte Subsets/virology , T-Lymphocytes, Helper-Inducer/virology , Virus Latency , Animals , CD4-Positive T-Lymphocytes/chemistry , Cell Differentiation , Immunophenotyping , Macaca mulatta , Programmed Cell Death 1 Receptor/analysis , Receptors, CXCR5/analysis , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/growth & development , T-Lymphocyte Subsets/chemistry , T-Lymphocytes, Helper-Inducer/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...