Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pathogens ; 13(2)2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38392915

RESUMO

Zika virus (ZIKV) is a reemerging flavivirus that is primarily spread through bites from infected mosquitos. It was first discovered in 1947 in sentinel monkeys in Uganda and has since been the cause of several outbreaks, primarily in tropical and subtropical areas. Unlike earlier outbreaks, the 2015-2016 epidemic in Brazil was characterized by the emergence of neurovirulent strains of ZIKV strains that could be sexually and perinatally transmitted, leading to the Congenital Zika Syndrome (CZS) in newborns, and Guillain-Barre Syndrome (GBS) along with encephalitis and meningitis in adults. The immune response elicited by ZIKV infection is highly effective and characterized by the induction of both ZIKV-specific neutralizing antibodies and robust effector CD8+ T cell responses. However, the structural similarities between ZIKV and Dengue virus (DENV) lead to the induction of cross-reactive immune responses that could potentially enhance subsequent DENV infection, which imposes a constraint on the development of a highly efficacious ZIKV vaccine. The isolation and characterization of antibodies capable of cross-neutralizing both ZIKV and DENV along with cross-reactive CD8+ T cell responses suggest that vaccine immunogens can be designed to overcome these constraints. Here we review the structural characteristics of ZIKV along with the evidence of neuropathogenesis associated with ZIKV infection and the complex nature of the immune response that is elicited by ZIKV infection.

2.
Nat Commun ; 14(1): 1264, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36882405

RESUMO

Human immunodeficiency virus (HIV) is a mucosally transmitted virus that causes immunodeficiency and AIDS. Developing efficacious vaccines to prevent infection is essential to control the epidemic. Protecting the vaginal and rectal mucosa, the primary routes of HIV entry has been a challenge given the significant compartmentalization between the mucosal and peripheral immune systems. We hypothesized that direct intranodal vaccination of mucosa associated lymphoid tissue (MALT) such as the readily accessible palatine tonsils could overcome this compartmentalization. Here we show that rhesus macaques primed with plasmid DNA encoding SIVmac251-env and gag genes followed by an intranodal tonsil MALT boost with MVA encoding the same genes protects from a repeated low dose intrarectal challenge with highly pathogenic SIVmac251; 43% (3/7) of vaccinated macaques remained uninfected after 9 challenges as compared to the unvaccinated control (0/6) animals. One vaccinated animal remained free of infection even after 22 challenges. Vaccination was associated with a ~2 log decrease in acute viremia that inversely correlated with anamnestic immune responses. Our results suggest that a combination of systemic and intranodal tonsil MALT vaccination could induce robust adaptive and innate immune responses leading to protection from mucosal infection with highly pathogenic HIV and rapidly control viral breakthroughs.


Assuntos
Infecções por HIV , Linfoma de Zona Marginal Tipo Células B , Vacínia , Animais , Humanos , Feminino , Tonsila Palatina , Macaca mulatta , Vaccinia virus , Vacinação
3.
Cell Immunol ; 366: 104396, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34157462

RESUMO

Human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infections are characterized by dramatic alterations in the mucosal CD4 T cell compartment. Though viremia is effectively suppressed, and peripheral CD4 T cell numbers recover to near healthy levels after highly active anti-retroviral therapy (HAART), some of the dysfunctional consequences of HIV infection continue to persist during therapy. We hypothesized that CD4 T follicular helper (Tfh) cell deficiencies may play a role in this process. Using the macaque model we show that SIV infection was associated with a significant loss of Tfh cells in the GALT that drain the mesentery lining the gastrointestinal tract (GIT). Loss of Tfh cells significantly correlated with the depletion of the overall memory CD4 T cell compartment; most Tfh cells in the GALT expressed a CD95+CD28+ memory phenotype suggesting that infection of the memory compartment likely drives the loss of GALT Tfh cells during infection. Continuous anti-retroviral therapy (cART) was accompanied by a significant repopulation of Tfh cells in the GALT to levels similar to those of uninfected animals. Repopulating Tfh cells displayed significantly higher capacity to produce IL-21 as compared to SIV infected animals suggesting that cART fully restores Tfh cells that are functionally capable of supporting GC reactions in the GALT.


Assuntos
Terapia Antirretroviral de Alta Atividade/métodos , Linfócitos T CD4-Positivos/imunologia , Centro Germinativo/imunologia , Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , Intestinos/imunologia , Tecido Linfoide/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia/fisiologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Movimento Celular , Modelos Animais de Doenças , Infecções por HIV/imunologia , Humanos , Memória Imunológica , Interleucinas/metabolismo , Linfopenia , Macaca , Receptor de Morte Celular Programada 1/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia
4.
Front Immunol ; 12: 657679, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33815419

RESUMO

Human Immunodeficiency Virus (HIV) and Simian Immunodeficiency Virus (SIV) are associated with severe perturbations in the gut mucosal environment characterized by massive viral replication and depletion of CD4 T cells leading to dysbiosis, breakdown of the epithelial barrier, microbial translocation, immune activation and disease progression. Multiple mechanisms play a role in maintaining homeostasis in the gut mucosa and protecting the integrity of the epithelial barrier. Among these are the secretory IgA (sIgA) that are produced daily in vast quantities throughout the mucosa and play a pivotal role in preventing commensal microbes from breaching the epithelial barrier. These microbe specific, high affinity IgA are produced by IgA+ plasma cells that are present within the Peyer's Patches, mesenteric lymph nodes and the isolated lymphoid follicles that are prevalent in the lamina propria of the gastrointestinal tract (GIT). Differentiation, maturation and class switching to IgA producing plasma cells requires help from T follicular helper (Tfh) cells that are present within these lymphoid tissues. HIV replication and CD4 T cell depletion is accompanied by severe dysregulation of Tfh cell responses that compromises the generation of mucosal IgA that in turn alters barrier integrity leading to commensal bacteria readily breaching the epithelial barrier and causing mucosal pathology. Here we review the effect of HIV infection on Tfh cells and mucosal IgA responses in the GIT and the consequences these have for gut dysbiosis and mucosal immunopathogenesis.


Assuntos
Microbioma Gastrointestinal/imunologia , Infecções por HIV/imunologia , HIV/imunologia , Homeostase/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunoglobulina A/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Comunicação Celular/imunologia , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Humanos , Imunidade nas Mucosas , Imunoglobulina A Secretora/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Plasmócitos/imunologia , Plasmócitos/metabolismo , Transdução de Sinais , Vírus da Imunodeficiência Símia/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
5.
J Radiat Res ; 62(2): 236-248, 2021 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-33616187

RESUMO

The lung is sensitive to radiation and exhibits several phases of injury, with an initial phase of radiation-induced pneumonitis followed by delayed and irreversible fibrosis. The angiotensin-converting enzyme inhibitor captopril has been demonstrated to mitigate radiation lung injury and to improve survival in animal models of thoracic irradiation, but the mechanism remains poorly understood. Here we investigated the effect of captopril on early inflammatory events in the lung in female CBA/J mice exposed to thoracic X-ray irradiation of 17-17.9 Gy (0.5-0.745 Gy min-1). For whole-body + thoracic irradiation, mice were exposed to 7.5 Gy (0.6 Gy min-1) total-body 60Co irradiation and 9.5 Gy thoracic irradiation. Captopril was administered orally (110 mg kg-1 day-1) in the drinking water, initiated 4 h through to150 days post-irradiation. Captopril treatment increased survival from thoracic irradiation to 75% at 150 days compared with 0% survival in vehicle-treated animals. Survival was characterized by a significant decrease in radiation-induced pneumonitis and fibrosis. Investigation of early inflammatory events showed that captopril significantly attenuated macrophage accumulation and decreased the synthesis of radiation-induced interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) pro-inflammatory cytokines in the lungs of irradiated mice. Suppression of IL-1ß and TNF-α correlated with an increase of the anti-inflammatory cytokine IL-10 in the spleen with captopril treatment. We also found that captopril decreased markers for radiation-induced accelerated senescence in the lung tissue. Our data suggest that suppression of inflammation and senescence markers, combined with an increase of anti-inflammatory factors, are a part of the mechanism for captopril-induced survival in thoracic irradiated mice.


Assuntos
Envelhecimento/patologia , Captopril/uso terapêutico , Pneumonia/tratamento farmacológico , Tórax/efeitos da radiação , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Biomarcadores/metabolismo , Captopril/farmacologia , Citocinas/metabolismo , Feminino , Mediadores da Inflamação/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/efeitos da radiação , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/patologia , Macrófagos Alveolares/efeitos da radiação , Camundongos Endogâmicos CBA , Fibrose Pulmonar/patologia , Baço/efeitos dos fármacos , Baço/efeitos da radiação , Análise de Sobrevida , Irradiação Corporal Total , Raios X
6.
Sci Rep ; 10(1): 6387, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286417

RESUMO

Human immunodeficiency virus (HIV) infection is characterized by a massive loss of CD4 T cells in the gastrointestinal tract (GIT) that is accompanied by changes in the gut microbiome and microbial translocation that contribute to inflammation and chronic immune activation. Though highly active antiretroviral therapy (HAART) has led to better long-term outcomes in HIV infected patients, it has not been as effective at reverting pathogenesis in the GIT. Using the simian immunodeficiency virus (SIV) infection model, we show that combination antiretroviral therapy (c-ART) partially reverted microbial dysbiosis observed during SIV infection. Though the relative abundance of bacteria, their richness or diversity did not significantly differ between infected and treated animals, microbial dysbiosis was evident via multiple beta diversity metrics: Jaccard similarity coefficient, Bray-Curtis similarity coefficient, and Yue & Clayton theta similarity coefficient. Principal coordinates analysis (PCoA) clustered SIV-infected untreated animals away from healthy and treated animals that were clustered closely, indicating that c-ART partially reversed the gut dysbiosis associated with SIV infection. Metastats analysis identified specific operational taxonomic units (OTUs) falling within the Streptococcus, Prevotella, Acinetobacter, Treponema, and Lactobacillus genera that were differentially represented across the three groups. Our results suggest that complete viral suppression with c-ART could potentially revert microbial dysbiosis observed during SIV and HIV infections.


Assuntos
Antirretrovirais/uso terapêutico , Disbiose/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/microbiologia , Animais , Bactérias/classificação , Bactérias/efeitos dos fármacos , Macaca mulatta , Vírus da Imunodeficiência Símia , Carga Viral/efeitos dos fármacos
7.
PLoS One ; 14(9): e0221159, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31490965

RESUMO

Gender differences in Human immunodeficiency virus (HIV) disease progression and comorbidities have been extensively reported. Using the simian immunodeficiency virus (SIV) infected rhesus macaque model, we show that these differences are apparent very early during the course of infection. Though there were no major changes in the proportions of CD4 T cells or its subsets, central memory CD4 T cells from female macaques were found to differentially regulate a significantly larger number of genes at day 4 post-infection (PI) as compared to males. Pathway analysis revealed divergence of both canonical and biological pathways that persisted at day 10 PI. Changes in gene expression profiles were accompanied by a significant increase in plasma levels of pro-inflammatory mediators such as MCP-1/CCL2, I-TAC/CXCL11, and MIF. Though plasma levels of IFNα did not differ between male and female macaques, the expression levels of IFNα subtype-14, 16, IFNß, and IFNω were significantly upregulated in the lymph nodes of female macaques at day 10 PI as compared to male macaques. Our results suggest that the pathogenic sequelae seen during chronic infection may be shaped by gender differences in immune responses induced very early during the course of HIV infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Perfilação da Expressão Gênica , Imunidade Inata , Caracteres Sexuais , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Doença Aguda , Animais , Linfócitos T CD4-Positivos/metabolismo , Quimiocina CXCL11/sangue , Feminino , Interferons/sangue , Macaca mulatta , Masculino , RNA Mensageiro/genética , Receptores CCR2/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/fisiopatologia
8.
Sci Rep ; 9(1): 7877, 2019 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-31133721

RESUMO

The consequences of simultaneous infection with Zika (ZIKV) and Dengue (DENV) viruses are poorly understood. Here we show that rhesus macaques experimentally coinfected simultaneously with ZIKV and DENV-2 demonstrated ZIKV or DENV replication without an enhancement of either infection. Coinfection was accompanied by an increase in the proportions of CD14+CD16+ pro-inflammatory subsets of monocytes and release of pro-inflammatory cytokines in the plasma. Numerous cytokines such as I-TAC, Eotaxin, RANTES, MCP-1, IFNγ and MIG demonstrated a biphasic peak that coincided with the differences in kinetics of ZIKV and DENV replication suggesting that viral replication likely differentially modulated the release of these cytokines. Red blood cell indices significantly declined during acute infection suggesting transient anemia, and was accompanied by elevated levels of muscle, liver and renal injury markers. These findings have implications for understanding the pathogenesis of coinfection in ZIKV and DENV endemic regions, and is the 1st report of an experimental coinfection using the rhesus macaque model for ZIKV and DENV infections.


Assuntos
Coinfecção/imunologia , Vírus da Dengue/imunologia , Dengue/imunologia , Macaca mulatta/virologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Coinfecção/sangue , Coinfecção/virologia , Citocinas/sangue , Citocinas/imunologia , Dengue/sangue , Dengue/virologia , Vírus da Dengue/fisiologia , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/sangue , Inflamação/imunologia , Inflamação/virologia , Macaca mulatta/sangue , Macaca mulatta/imunologia , Masculino , Monócitos/imunologia , Monócitos/virologia , Carga Viral , Replicação Viral , Zika virus/fisiologia , Infecção por Zika virus/sangue , Infecção por Zika virus/virologia
9.
J Comp Neurol ; 527(10): 1706-1719, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30680733

RESUMO

Although initial observations of infections with the Zika virus describe a mild illness, more recent reports show that infections by Zika result in neurotropism. In 2015, substantial congenital malformations were observed, with numerous infants born with microcephaly in Brazil. To study the underlying mechanism and effects of the disease, it is critical to find suitable animal models. Rodents lack an immune system parallel to humans and also have lissencephalic brains, which are likely to react differently to infections. As the smallest gyrencephalic mammal, ferrets may provide an important animal model to study the Zika virus, as their brains share many characteristics with humans. To evaluate the prospect of using ferrets to study Zika virus infection, we injected seven pregnant jills with the PR strain subcutaneously on gestational day 21, corresponding to the initiation of corticogenesis. These injections resulted in mixed effects. Two animals died of apparent infection, and all kits were resorbed in another animal that did not die. The other four animals remained pregnant until gestational day 40, when the kits were delivered by caesarian section. We evaluated the animals using CT, MRI, diffusion tensor imaging, and immunohistochemistry. The kits displayed a number of features compatible with an infection that impacted both the brain and skull. The outcomes, however, were variable and differed within and across litters, which ranged from the absence of observable abnormalities to prominent changes, suggesting differential vulnerability of kits to infection by the Zika virus or to subsequent mechanisms of neurodevelopmental disruption.


Assuntos
Encéfalo/patologia , Modelos Animais de Doenças , Infecção por Zika virus/patologia , Animais , Animais Recém-Nascidos , Furões
10.
J Cell Mol Med ; 22(11): 5682-5687, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30191661

RESUMO

Human immunodeficiency virus (HIV) infection is characterized by dysfunctional B cell responses. Here we show that chronic simian immunodeficiency virus (SIV) infection is characterized by an expansion of either lymph node germinal center (GC) B cells that co-express Bcl6, Ki-67 and IL-21R and correlate with expanded T follicular helper (Tfh) cells or B cells that lack Bcl6, Ki-67 and IL-21R but express high levels of anti-apoptotic Bcl2 that negatively correlate with Tfh cells. The lack of Tfh cells likely contributes to persistence of dysfunctional non-proliferating B cells during chronic infection. These findings have implications for protective immunity in HIV-infected individuals who harbour low frequencies of Tfh cells.


Assuntos
Linfócitos B/imunologia , Infecções por HIV/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/genética , Animais , Linfócitos B/patologia , Diferenciação Celular/imunologia , Centro Germinativo/imunologia , Centro Germinativo/virologia , Infecções por HIV/genética , Infecções por HIV/patologia , Infecções por HIV/virologia , Humanos , Linfonodos/imunologia , Linfonodos/patologia , Macaca mulatta/imunologia , Fenótipo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-6/genética , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/patogenicidade , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/patologia
11.
J Cell Mol Med ; 22(9): 4274-4282, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29971909

RESUMO

Allogeneic stem cell transplantation is currently the only curative therapy for primary myelofibrosis (MF), while the JAK2 inhibitor, ruxolitinib. Has been approved only for palliation. Other therapies are desperately needed to reverse life-threatening MF. However, the cell(s) and cytokine(s) that promote MF remain unclear. Several reports have demonstrated that captopril, an inhibitor of angiotensin-converting enzyme that blocks the production of angiotensin II (Ang II), mitigates fibrosis in heart, lung, skin and kidney. Here, we show that captopril can mitigate the development of MF in the Gata1low mouse model of primary MF. Gata1low mice were treated with 79 mg/kg/d captopril in the drinking water from 10 to 12 months of age. At 13 months of age, bone marrows were examined for fibrosis, megakaryocytosis and collagen expression; spleens were examined for megakaryocytosis, splenomegaly and collagen expression. Treatment of Gata1low mice with captopril in the drinking water was associated with normalization of the bone marrow cellularity; reduced reticulin fibres, splenomegaly and megakaryocytosis; and decreased collagen expression. Our findings suggest that treating with the ACE inhibitors captopril has a significant benefit in overcoming pathological changes associated with MF.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Antineoplásicos/farmacologia , Captopril/farmacologia , Fator de Transcrição GATA1/genética , Mielofibrose Primária/tratamento farmacológico , Esplenomegalia/tratamento farmacológico , Administração Oral , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Colágeno/antagonistas & inibidores , Colágeno/genética , Colágeno/metabolismo , Modelos Animais de Doenças , Água Potável/administração & dosagem , Reposicionamento de Medicamentos , Feminino , Fator de Transcrição GATA1/deficiência , Expressão Gênica , Masculino , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Megacariócitos/patologia , Camundongos , Camundongos Knockout , Mielofibrose Primária/genética , Mielofibrose Primária/metabolismo , Mielofibrose Primária/patologia , Reticulina/antagonistas & inibidores , Reticulina/genética , Reticulina/metabolismo , Esplenomegalia/genética , Esplenomegalia/metabolismo , Esplenomegalia/patologia
12.
Emerg Microbes Infect ; 7(1): 130, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-30006514

RESUMO

Structural similarities between Zika (ZIKV) and dengue virus (DENV) leads to the induction of cross-reactive responses. We have previously demonstrated that ZIKV exposed macaques significantly enhance DENV viremia. Here we show that this enhancement of DENV infection occurred in the presence of high levels of DENV cross-reactive IgG1 subclass of binding antibodies (bAb) with low DENV neutralizing antibody (nAb) activity (<1:10). The DENV-2 nAb titres after ZIKV infection were, however, higher than those induced in DENV-2 only infected animals suggesting that ZIKV induced low titres of cross-nAb against DENV. Surprisingly, DENV-2 infection of animals previously infected with ZIKV was not accompanied by an anamnestic increase in cross-nAb titres till about 1 week after DENV-2 infection. This delay coincided with enhanced DENV-2 viremia indicating that high levels of cross-bAb in the absence of high nAb contributes to enhancement of DENV infection. Serum collected 8 weeks after DENV-2 infection had high levels of nAb and showed delayed antibody dependent enhancement (ADE) of infection (1:100 dilution) as compared with serum that was collected from ZIKV infected animals prior to DENV-2 infection (1:10 dilution). Examination of serum from macaques that were simultaneously infected with both ZIKV and DENV-2 showed high levels of nAb and delayed ADE responses raising the possibility that the low levels of cross-nAb induced by ZIKV infection could be overcome by co-immunization against ZIKV and DENV infection. Taken together, our results provide additional insights into the nature and kinetics of cross-reactive antibody responses and identify a critical correlate that could potentially prevent enhancement of DENV infection during ZIKV convalescence.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vírus da Dengue/imunologia , Dengue/imunologia , Macaca/imunologia , Macaca/virologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Formação de Anticorpos , Anticorpos Facilitadores , Coinfecção/imunologia , Coinfecção/virologia , Reações Cruzadas/imunologia , Dengue/virologia , Modelos Animais de Doenças , Imunoglobulina G/imunologia , Testes de Neutralização , Infecção por Zika virus/virologia
13.
Open Forum Infect Dis ; 5(7): ofy151, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30019003

RESUMO

Zika virus infection in a dengue virus-naïve subject was associated with the induction of high levels of cross-reactive binding antibodies. These responses were, however, largely non-neutralizing and displayed a capacity to enhance dengue infection in vitro at significantly low dilution (1:10). In contrast, a subject who had high levels of neutralizing antibodies against both dengue and Zika viruses enhanced infection at a dilution of 1:10 000. These results suggest that high levels of dengue cross-neutralizing antibodies could potentially prevent the enhancement of dengue infection in Zika virus-convalescent individuals.

14.
J Vis Exp ; (134)2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29708542

RESUMO

Antibody dependent enhancement of infection has been shown to play a major role in Dengue viral pathogenesis. Traditional assays that measure the capacity of antibodies or serum to enhance infection in impermissible cell lines have relied on using viral output in the media followed by plaque assays to quantify infection. More recently, these assays have examined Dengue virus (DENV) infection in the cell lines using fluorescently labeled antibodies. Both these approaches have limitations that restrict the widespread use of these techniques. Here, we describe a simple in vitro assay using Dengue virus reporter viral particles (RVPs) that express green fluorescent protein and K562 cells to examine antibody dependent enhancement (ADE) of DENV infection using serum that was obtained from rhesus macaques 16 weeks after infection with Zika virus (ZIKV). This technique is reliable, involves minimal manipulation of cells, does not involve the use of live replication competent virus, and can be performed in a high throughput format to get a quantitative readout using flow cytometry. Additionally, this assay can be easily adapted to examine antibody dependent enhancement (ADE) of other flavivirus infections such as Yellow Fever virus (YFV), Japanese Equine Encephalitis virus (JEEV), West Nile virus (WNV) etc. where RVPs are available. The ease of setting up the assay, analyzing the data, and interpreting results makes it highly amenable to most laboratory settings.


Assuntos
Anticorpos Facilitadores/genética , Vírus da Dengue/patogenicidade , Citometria de Fluxo/métodos , Zika virus/patogenicidade , Animais , Humanos
15.
Front Immunol ; 9: 299, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29520278

RESUMO

Human immunodeficiency virus (HIV) establishes life-long latency in infected individuals. Although highly active antiretroviral therapy (HAART) has had a significant impact on the course of HIV infection leading to a better long-term outcome, the pool of latent reservoir remains substantial even under HAART. Numerous approaches have been under development with the goal of eradicating the latent HIV reservoir though with limited success. Approaches that combine immune-mediated control of HIV to activate both the innate and the adaptive immune system under suppressive therapy along with "shock and kill" drugs may lead to a better control of the reactivated virus. Interferon-α (IFN-α) is an innate cytokine that has been shown to activate intracellular defenses capable of restricting and controlling HIV. IFN-α, however, harbors numerous functional subtypes that have been reported to display different binding affinities and potency. Recent studies have suggested that certain subtypes such as IFN-α8 and IFN-α14 have potent anti-HIV activity with little or no immune activation, whereas other subtypes such as IFN-α4, IFN-α5, and IFN-α14 activate NK cells. Could these subtypes be used in combination with other strategies to reduce the latent viral reservoir? Here, we review the role of IFN-α subtypes in HIV infection and discuss the possibility that certain subtypes could be potential adjuncts to a "shock and kill" or therapeutic vaccination strategy leading to better control of the latent reservoir and subsequent functional cure.


Assuntos
Vacinas contra a AIDS/farmacologia , Fármacos Anti-HIV/farmacologia , Infecções por HIV/tratamento farmacológico , Interferon-alfa/uso terapêutico , Latência Viral/efeitos dos fármacos , Humanos
16.
Sci Rep ; 7(1): 10498, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28874759

RESUMO

Structural and functional homologies between the Zika and Dengue viruses' envelope proteins raise the possibility that cross-reactive antibodies induced following Zika virus infection might enhance subsequent Dengue infection. Using the rhesus macaque model we show that prior infection with Zika virus leads to a significant enhancement of Dengue-2 viremia that is accompanied by neutropenia, lympocytosis, hyperglycemia, and higher reticulocyte counts, along with the activation of pro-inflammatory monocyte subsets and release of inflammatory mediators. Zika virus infection induced detectable Dengue cross-reactive serum IgG responses that significantly amplified after Dengue-2 virus infection. Serum from Zika virus immune animals collected prior to Dengue-2 infection showed significant capacity for in vitro antibody dependent enhancement of Dengue-1, 2, 3 and 4 serotypes suggesting that pre-existing immunity to Zika virus could potentially enhance infection by heterologous Dengue serotypes. Our results provide first in vivo evidence that prior exposure to Zika virus infection can enhance Dengue infection, which has implications for understanding pathogenesis and the development of vaccines.


Assuntos
Coinfecção , Vírus da Dengue/fisiologia , Dengue/veterinária , Doenças dos Macacos/virologia , Viremia , Infecção por Zika virus/veterinária , Zika virus/fisiologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Facilitadores/imunologia , Biologia Computacional/métodos , Reações Cruzadas/imunologia , Citocinas/metabolismo , Vírus da Dengue/classificação , Mediadores da Inflamação/metabolismo , Macaca mulatta , Doenças dos Macacos/imunologia , Testes de Neutralização , Carga Viral
17.
Cell Immunol ; 310: 156-164, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27622386

RESUMO

Innate interferons (IFN) are comprised of multiple Type I and III subtypes. The in vivo kinetics of subtype responses during human immunodeficiency virus (HIV) infection is not well defined. Using the acute simian immunodeficiency virus (SIV) infection model, we show that plasma IFNα levels peak at day 10 post-infection (pi) after which they rapidly declined. The mRNA expression of Type I and III IFN subtypes were significantly elevated in the lymph nodes (LN) at day 10 pi. Though the expression levels of all subtypes declined by day 14-31 pi, numerous subtypes remained elevated suggesting that ongoing viral replication in LN continues to drive induction of these subtypes. Interestingly, treatment with reverse transcriptase (RT) inhibitors at day 7 pi significantly suppressed plasma IFNα responses by day 10 pi that significantly correlated with cell-associated SIV DNA loads suggesting that RT byproducts such as viral DNA likely plays a role in driving IFN responses during acute SIV infection. Quantification of Type I and III subtype transcripts in sorted subsets of LN CD4+ and CD8+ T cells, CD14+/CD14- monocytes/macrophages, and total CD11c/CD123+ dendritic cells (DC) at day 10 pi showed that DC expressed ∼3-4 log more subtype transcripts as compared to the other subsets. Taken together, our results provide new insights into the kinetics of innate interferon responses during early stages of infection, and provide evidence that DC's are a major in vivo source of innate IFN during acute SIV infection.


Assuntos
Síndrome da Imunodeficiência Adquirida/terapia , Células Dendríticas/efeitos dos fármacos , Interferon-alfa/biossíntese , Linfonodos/imunologia , Inibidores da Transcriptase Reversa/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia/imunologia , Síndrome da Imunodeficiência Adquirida/imunologia , Doença Aguda , Animais , Células Cultivadas , DNA Viral/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Terapia de Imunossupressão , Interferon-alfa/sangue , Linfonodos/virologia , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia
18.
J Immunol Res ; 2015: 673815, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26065003

RESUMO

Human and simian immunodeficiency virus (HIV and SIV) infections are characterized by manifestation of numerous opportunistic infections and inflammatory conditions in the oral mucosa. The loss of CD4(+) T cells that play a critical role in maintaining mucosal immunity likely contributes to this process. Here we show that CD4(+) T cells constitute a minor population of T cells in the oral mucosa and display a predominantly central memory phenotype mirroring other mucosal sites such as the rectal mucosa. Chronic SIV infection was associated with a near total depletion of CD4(+) T cells in the oral mucosa that appear to repopulate during antiretroviral therapy (ART). Repopulating CD4(+) T cells harbored a large fraction of Th17 cells suggesting that ART potentially reconstitutes oral mucosal immunity. However, a minor fraction of repopulating CD4(+) T cells harbored SIV DNA suggesting that the viral reservoir continues to persist in the oral mucosa during ART. Therapeutic approaches aimed at obtaining sustainable CD4(+) T cell repopulation in combination with strategies that can eradicate the latent viral reservoir in the oral mucosa are essential for better oral health and long-term outcome in HIV infected patients.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Mucosa Bucal/imunologia , Mucosa Bucal/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Terapia Antirretroviral de Alta Atividade/métodos , DNA Viral/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Imunidade nas Mucosas/imunologia , Memória Imunológica/imunologia , Mucosa Intestinal/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia/efeitos dos fármacos
19.
J Virol ; 89(5): 2972-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25540365

RESUMO

Chronic human immunodeficiency virus and simian immunodeficiency virus (HIV and SIV) infections are characterized by mucosal inflammation in the presence of anti-inflammatory cytokines such as transforming growth factor ß (TGFß). The mechanisms for refractiveness to TGFß are not clear. Here we show that the expression of microRNA miR-155 was significantly upregulated in the oropharyngeal mucosa during chronic SIV infection and was coincident with downregulation of TGFß receptor 2 (TGFß-R2) and SMAD5, key TGFß signaling genes that harbor putative target sites for miR-155. Ectopic expression of miR-155 in vitro was found to significantly downregulate TGFß-R2 and Smad5 expression, suggesting a role for miR-155 in the suppression of TGFß-R2 and SMAD5 genes in vivo. The downregulation of TGFß signaling genes by miR-155 likely contributes to the nonresponsiveness to TGFß during SIV infection and may inadvertently aid in increased immune activation during HIV and SIV infections.


Assuntos
Interações Hospedeiro-Patógeno , MicroRNAs/genética , Mucosa Bucal/patologia , Receptores de Fatores de Crescimento Transformadores beta/genética , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia/fisiologia , Proteína Smad5/biossíntese , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Macaca mulatta , Orofaringe/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia
20.
Clin Vaccine Immunol ; 21(11): 1469-73, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25185575

RESUMO

The gastrointestinal tract (GIT) is a primary site for human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infection, replication, and dissemination. After an initial explosive phase of infection, HIV establishes latency. In addition to CD4 T cells, macrophages are readily infected, which can persist for long periods of time. Though macrophages at various systemic sites are infected, those present in the GIT constitute a major cellular reservoir due to the abundance of these cells at mucosal sites. Here, we review some of the important findings regarding what is known about the macrophage reservoir in the gut and explore potential approaches being pursued in the field to reduce this reservoir. The development of strategies that can lead to a functional cure will need to incorporate approaches that can eradicate the macrophage reservoir in the GIT.


Assuntos
Trato Gastrointestinal/imunologia , Trato Gastrointestinal/virologia , Infecções por HIV/virologia , HIV/fisiologia , Macrófagos/virologia , Latência Viral , Replicação Viral , Antivirais/farmacologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...