Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Trop Med Infect Dis ; 6(3)2021 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-34287349

RESUMO

The antigenic diversity of Orientia tsutsugamushi as well as the interstrain difference(s) associated with virulence in mice impose the necessity to dissect the host immune response. In this study we compared the host response in lethal and non-lethal murine models of O. tsutsugamushi infection using the two strains, Karp (New Guinea) and Woods (Australia). The models included the lethal model: Karp intraperitoneal (IP) challenge; and the nonlethal models: Karp intradermal (ID), Woods IP, and Woods ID challenges. We monitored bacterial trafficking to the liver, lung, spleen, kidney, heart, and blood, and seroconversion during the 21-day challenge. Bacterial trafficking to all organs was observed in both the lethal and nonlethal models of infection, with significant increases in average bacterial loads observed in the livers and hearts of the lethal model. Multicolor flow cytometry was utilized to analyze the CD4+ and CD8+ T cell populations and their intracellular production of the cytokines IFNγ, TNF, and IL2 (single, double, and triple combinations) associated with both the lethal and nonlethal murine models of infection. The lethal model was defined by a cytokine signature of double- (IFNγ-IL2) and triple-producing (IL2-TNF-IFNγ) CD4+ T-cell populations; no multifunctional signature was identified in the CD8+ T-cell populations associated with the lethal model. In the nonlethal model, the cytokine signature was predominated by CD4+ and CD8+ T-cell populations associated with single (IL2) and/or double (IL2-TNF) populations of producers. The cytokine signatures associated with our lethal model will become depletion targets in future experiments; those signatures associated with our nonlethal model are hypothesized to be related to the protective nature of the nonlethal challenges.

2.
PLoS Negl Trop Dis ; 15(7): e0009559, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34314415

RESUMO

Viral and parasitic coinfections are known to lead to both enhanced disease progression and altered disease states. HTLV-1 and Strongyloides stercoralis are co-endemic throughout much of their worldwide ranges resulting in a significant incidence of coinfection. Independently, HTLV-1 induces a Th1 response and S. stercoralis infection induces a Th2 response. However, coinfection with the two pathogens has been associated with the development of S. stercoralis hyperinfection and an alteration of the Th1/Th2 balance. In this study, a model of HTLV-1 and S. stercoralis coinfection in CD34+ umbilical cord blood hematopoietic stem cell engrafted humanized mice was established. An increased level of mortality was observed in the HTLV-1 and coinfected animals when compared to the S. stercoralis infected group. The mortality was not correlated with proviral loads or total viral RNA. Analysis of cytokine profiles showed a distinct shift towards Th1 responses in HTLV-1 infected animals, a shift towards Th2 cytokines in S. stercoralis infected animals and elevated TNF-α responses in coinfected animals. HTLV-1 infected and coinfection groups showed a significant, yet non-clonal expansion of the CD4+CD25+ T-cell population. Numbers of worms in the coinfection group did not differ from those of the S. stercoralis infected group and no autoinfective larvae were found. However, infective larvae recovered from the coinfection group showed an enhancement in growth, as was seen in mice with S. stercoralis hyperinfection caused by treatment with steroids. Humanized mice coinfected with S. stercoralis and HTLV-1 demonstrate features associated with human infection with these pathogens and provide a unique opportunity to study the interaction between these two infections in vivo in the context of human immune cells.


Assuntos
Antígenos CD34/sangue , Citocinas/metabolismo , Infecções por HTLV-I/imunologia , Células-Tronco Hematopoéticas/metabolismo , Strongyloides stercoralis/crescimento & desenvolvimento , Estrongiloidíase/imunologia , Animais , Linhagem Celular , Coinfecção , Citocinas/genética , Sangue Fetal , Infecções por HTLV-I/complicações , Vírus Linfotrópico T Tipo 1 Humano , Larva/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estrongiloidíase/complicações
3.
PLoS Negl Trop Dis ; 13(9): e0007730, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31525197

RESUMO

BACKGROUND: The current strategy for the elimination of onchocerciasis is based on annual or bi-annual mass drug administration with ivermectin. However, due to several limiting factors there is a growing concern that elimination of onchocerciasis cannot be achieved solely through the current strategy. Additional tools are critically needed including a prophylactic vaccine. Presently Ov-103 and Ov-RAL-2 are the most promising vaccine candidates against an Onchocerca volvulus infection. METHODOLOGY/PRINCIPAL FINDINGS: Protection induced by immunization of mice with the alum-adjuvanted Ov-103 or Ov-RAL-2 vaccines appeared to be antibody dependent since AID-/- mice that could not mount antigen-specific IgG antibody responses were not protected from an Onchocerca volvulus challenge. To determine a possible association between antigen-specific antibody responses and anti-larvae protective immunity in humans, we analyzed the presence of anti-Ov-103 and anti-Ov-RAL-2 cytophilic antibody responses (IgG1 and IgG3) in individuals classified as putatively immune, and in infected individuals who developed concomitant immunity with age. It was determined that 86% of putatively immune individuals and 95% individuals with concomitant immunity had elevated IgG1 and IgG3 responses to Ov-103 and Ov-RAL-2. Based on the elevated chemokine levels associated with protection in the Ov-103 or Ov-RAL-2 immunized mice, the profile of these chemokines was also analyzed in putatively immune and infected individuals; both groups contained significantly higher levels of KC, IP-10, MCP-1 and MIP-1ß in comparison to normal human sera. Moreover, human monospecific anti-Ov-103 antibodies but not anti-Ov-RAL-2 significantly inhibited the molting of third-stage larvae (L3) in vitro by 46% in the presence of naïve human neutrophils, while both anti-Ov-103 and anti-Ov-RAL-2 antibodies significantly inhibited the molting by 70-80% when cultured in the presence of naive human monocytes. Interestingly, inhibition of molting by Ov-103 antibodies and monocytes was only in part dependent on contact with the cells, while inhibition of molting with Ov-RAL-2 antibodies was completely dependent on contact with the monocytes. In comparison, significant levels of parasite killing in Ov-103 and Ov-RAL-2 vaccinated mice only occurred when cells enter the parasite microenvironment. Taken together, antibodies to Ov-103 and Ov-RAL-2 and cells are required for protection in mice as well as for the development of immunity in humans. CONCLUSIONS/SIGNIFICANCE: Alum-adjuvanted Ov-103 and Ov-RAL-2 vaccines have the potential of reducing infection and thus morbidity associated with onchocerciasis in humans. The development of cytophilic antibodies, that function in antibody-dependent cellular cytotoxicity, is essential for a successful prophylactic vaccine against this infection.


Assuntos
Imunogenicidade da Vacina , Onchocerca volvulus/imunologia , Oncocercose/imunologia , Vacinas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen , Animais , Anticorpos Anti-Helmínticos/imunologia , Antígenos de Helmintos/imunologia , Quimiocinas/sangue , Imunoglobulina G/sangue , Larva/crescimento & desenvolvimento , Larva/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos , Onchocerca volvulus/crescimento & desenvolvimento , Oncocercose/parasitologia , Oncocercose/prevenção & controle , Vacinação , Vacinas/administração & dosagem
4.
PLoS Negl Trop Dis ; 12(12): e0006977, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30540742

RESUMO

BACKGROUND: The study of Onchocerca volvulus has been limited by its host range, with only humans and non-human primates shown to be susceptible to the full life cycle infection. Small animal models that support the development of adult parasites have not been identified. METHODOLOGY/PRINCIPAL FINDINGS: We hypothesized that highly immunodeficient NSG mice would support the survival and maturation of O. volvulus and alteration of the host microenvironment through the addition of various human cells and tissues would further enhance the level of parasite maturation. NSG mice were humanized with: (1) umbilical cord derived CD34+ stem cells, (2) fetal derived liver, thymus and CD34+ stem cells or (3) primary human skeletal muscle cells. NSG and humanized NSG mice were infected with 100 O. volvulus infective larvae (L3) for 4 to 12 weeks. When necropsies of infected animals were performed, it was observed that parasites survived and developed throughout the infection time course. In each of the different humanized mouse models, worms matured from L3 to advanced fourth stage larvae, with both male and female organ development. In addition, worms increased in length by up to 4-fold. Serum and urine, collected from humanized mice for identification of potential biomarkers of infection, allowed for the identification of 10 O. volvulus-derived proteins found specifically in either the urine or the serum of the humanized O. volvulus-infected NSG mice. CONCLUSIONS/SIGNIFICANCE: The newly identified mouse models for onchocerciasis will enable the development of O. volvulus specific biomarkers, screening for new therapeutic approaches and potentially studying the human immune response to infection with O. volvulus.


Assuntos
Biomarcadores/sangue , Biomarcadores/urina , Proteínas de Helminto/sangue , Proteínas de Helminto/urina , Onchocerca volvulus/crescimento & desenvolvimento , Oncocercose/diagnóstico , Animais , Modelos Animais de Doenças , Humanos , Estágios do Ciclo de Vida , Camundongos , Camundongos Endogâmicos NOD , Onchocerca volvulus/isolamento & purificação , Onchocerca volvulus/fisiologia , Oncocercose/sangue , Oncocercose/parasitologia , Oncocercose/urina
5.
Proc Natl Acad Sci U S A ; 115(1): 204-209, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29203662

RESUMO

Strongyloides stercoralis hyperinfection causes high mortality rates in humans, and, while hyperinfection can be induced by immunosuppressive glucocorticoids, the pathogenesis remains unknown. Since immunocompetent mice are resistant to infection with S. stercoralis, we hypothesized that NSG mice, which have a reduced innate immune response and lack adaptive immunity, would be susceptible to the infection and develop hyperinfection. Interestingly, despite the presence of large numbers of adult and first-stage larvae in S. stercoralis-infected NSG mice, no hyperinfection was observed even when the mice were treated with a monoclonal antibody to eliminate residual granulocyte activity. NSG mice were then infected with third-stage larvae and treated for 6 wk with methylprednisolone acetate (MPA), a synthetic glucocorticoid. MPA treatment of infected mice resulted in 50% mortality and caused a significant >10-fold increase in the number of parasitic female worms compared with infected untreated mice. In addition, autoinfective third-stage larvae, which initiate hyperinfection, were found in high numbers in MPA-treated, but not untreated, mice. Remarkably, treatment with Δ7-dafachronic acid, an agonist of the parasite nuclear receptor Ss-DAF-12, significantly reduced the worm burden in MPA-treated mice undergoing hyperinfection with S. stercoralis Overall, this study provides a useful mouse model for S. stercoralis autoinfection and suggests a therapeutic strategy for treating lethal hyperinfection.


Assuntos
Colestenos/farmacologia , Metilprednisolona/análogos & derivados , Strongyloides stercoralis/imunologia , Estrongiloidíase/tratamento farmacológico , Estrongiloidíase/imunologia , Animais , Colestenos/efeitos adversos , Feminino , Metilprednisolona/efeitos adversos , Metilprednisolona/farmacologia , Acetato de Metilprednisolona , Camundongos , Estrongiloidíase/patologia
6.
PLoS Negl Trop Dis ; 10(7): e0004797, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27387453

RESUMO

BACKGROUND: In some regions in Africa, elimination of onchocerciasis may be possible with mass drug administration, although there is concern based on several factors that onchocerciasis cannot be eliminated solely through this approach. A vaccine against Onchocerca volvulus would provide a critical tool for the ultimate elimination of this infection. Previous studies have demonstrated that immunization of mice with Ov-103 and Ov-RAL-2, when formulated with alum, induced protective immunity. It was hypothesized that the levels of protective immunity induced with the two recombinant antigens formulated with alum would be improved by formulation with other adjuvants known to enhance different types of antigen-specific immune responses. METHODOLOGY/ PRINCIPAL FINDINGS: Immunizing mice with Ov-103 and Ov-RAL-2 in conjunction with alum, Advax 2 and MF59 induced significant levels of larval killing and host protection. The immune response was biased towards Th2 with all three of the adjuvants, with IgG1 the dominant antibody. Improved larval killing and host protection was observed in mice immunized with co-administered Ov-103 and Ov-RAL-2 in conjunction with each of the three adjuvants as compared to single immunizations. Antigen-specific antibody titers were significantly increased in mice immunized concurrently with the two antigens. Based on chemokine levels, it appears that neutrophils and eosinophils participate in the protective immune response induced by Ov-103, and macrophages and neutrophils participate in immunity induced by Ov-RAL-2. CONCLUSIONS/SIGNIFICANCE: The mechanism of protective immunity induced by Ov-103 and Ov-RAL-2, with the adjuvants alum, Advax 2 and MF59, appears to be multifactorial with roles for cytokines, chemokines, antibody and specific effector cells. The vaccines developed in this study have the potential of reducing the morbidity associated with onchocerciasis in humans.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos de Helmintos/imunologia , Onchocerca volvulus/imunologia , Oncocercose/imunologia , Vacinas/imunologia , Animais , Anticorpos Anti-Helmínticos/imunologia , Antígenos de Helmintos/administração & dosagem , Antígenos de Helmintos/genética , Citocinas/imunologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Onchocerca volvulus/genética , Oncocercose/parasitologia , Oncocercose/prevenção & controle , Vacinação , Vacinas/administração & dosagem , Vacinas/genética
7.
Transgenic Res ; 22(1): 179-85, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22767020

RESUMO

The NOD.Cg-Prkdc ( scid ) Il2rg ( tm1Wjl )/SzJ mouse strain, commonly known as NSG (for NOD SCID Gamma) is severely immunodeficient and thus is an excellent recipient for xenografts, and in particular for engrafting human tumor cells and human hematopoietic stem cells. In the latter case, these cells give rise to many human hematopoetic lineages in their NSG hosts, resulting in recapitulation of many of the features of a human immune system. However, the immune system of these "humanized mice" (huMice) is not completely functional, in part because of a lack of expression of necessary human cytokines and HLA molecules by NSG host tissues. In order to facilitate the genetic modification of this strain in order to improve the huMouse model, we have created germline competent ES cells of this strain in which such modifications can be carried out.


Assuntos
Linhagem Celular/citologia , Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas , Animais , Linhagem Celular/imunologia , Linhagem da Célula/imunologia , Células-Tronco Embrionárias/imunologia , Células Germinativas/citologia , Células Germinativas/imunologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/imunologia , Camundongos , Camundongos SCID , Transplante Heterólogo
8.
Virus Res ; 163(2): 461-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22119401

RESUMO

The cytokine interleukin-1 beta (IL-1ß) is a potent inflammatory mediator in response to infection, and can be used as an immunological adjuvant. In this study, we constructed a recombinant porcine reproductive and respiratory syndrome virus (vP129/swIL1ß) expressing swine IL-1ß from the separate subgenomic mRNA inserted between the ORF1b and ORF2 genome region. MARC-145 cells infected with vP129/swIL1ß secreted 1947 pg of IL-1ß per 2 × 10(5)cells at 36 h post-infection. In vitro growth kinetics analysis in MARC-145 cells showed that the vP129/swIL1ß virus had a similar replication rate as that of parental virus. We further performed in vivo characterization of the vP129/swIL1ß virus in a nursery pig disease model. The vP129/swIL1ß infected pigs did not show visible clinical signs, while respiratory distress and lethargy were evident in pigs infected with the parental virus. The expression of various cytokines from peripheral blood mononuclear cells measured by fluorescent microsphere immunoassay showed that IL-1ß, IL-4 and IFN-γ expression levels were up-regulated in pigs infected with vP129/swIL1ß at 7 and 14 days post-infection. However, no detectable level of IL-1ß was found in serum samples from pigs infected with either vP129/swIL1ß or parental virus. In summary, this study demonstrated a recombinant PRRSV as a useful tool to study the role of different cytokines in disease progression and immune responses, which represents a new strategy for future therapeutic application and vaccine development.


Assuntos
Expressão Gênica , Interleucina-1beta/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Animais , Linhagem Celular , Citocinas/biossíntese , Interleucina-1beta/genética , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/patologia , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/crescimento & desenvolvimento , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Suínos , Carga Viral , Replicação Viral
9.
Intervirology ; 54(6): 339-48, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21293096

RESUMO

OBJECTIVES: Bile acids promoted the replication of hepatitis C virus (HCV) and compromised the anti-HCV effects of interferon-α (IFN-α) in replicon-harboring cells. To explore a potential mechanism for the observation, we studied the effects of bile acids on the epidermal growth factor receptor (EGFR) and the extracellular signal-regulated kinase (ERK) pathway in association with HCV replication in genotype 1a or 1b replicon-harboring cells. METHODS: Replicon-harboring cells were treated with various bile acids, IFN-α and small molecule inhibitors either individually or combined together. The effects of these treatments were measured using cell cycle analysis, qRT-PCR, and Western blot analysis. RESULTS: Bile acids induced the activation of EGFR/ERK pathway and extended S-phase of cells, which was correlated with the increased levels of viral replication. The inhibitors of EGFR (AG1478) or ERK (U0126) significantly mitigated the bile acid-mediated promotion of HCV replication. When AG1478 or U0126 were added to the treatment of bile acids and IFN-α, they were able to restore the anti-HCV effects of IFN-α. CONCLUSION: Our data suggest that the addition of an EGFR or ERK inhibitor to the current IFN-α-based regimen may improve overall treatment efficacy by blocking the bile acid-mediated promotion of HCV replication.


Assuntos
Ácidos e Sais Biliares/farmacologia , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hepacivirus/fisiologia , Replicação Viral , Antivirais/farmacologia , Ácidos e Sais Biliares/fisiologia , Ciclo Celular , Linhagem Celular , Ácido Quenodesoxicólico/farmacologia , Receptores ErbB/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Genes Reporter , Hepatite C/tratamento farmacológico , Humanos , Interferon-alfa/farmacologia , Interfase , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Regiões Promotoras Genéticas , Quinazolinas/farmacologia , Ativação Transcricional , Tirfostinas/farmacologia , Proteínas não Estruturais Virais/metabolismo
10.
Virus Res ; 140(1-2): 161-71, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19111584

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) has a specific cell tropism for differentiated macrophages, such as porcine alveolar macrophages (PAMs). We analyzed the expression of CD163 on PAMs and macrophages derived from CD14 positive blood monocytes (MDMs), in correlation with PRRSV replication. By flow cytometry analysis, we showed that the levels of CD163 expression correlated well with the overall level of PRRSV replication. We further examined the effects of modulators of macrophage function, including 12-O-tetradecanoylphorbol-13-acetate (TPA), lipopolysaccharide (LPS), and interleukin (IL)-10 on the expression of CD163 and PRRSV replication. Pre-treatment of PAMs or MDMs with TPA or LPS resulted in decreased expression of CD163 and reduction in PRRSV replication. On the contrary, the incubation of CD14 positive monocytes with IL-10 during differentiation into MDMs resulted in up-regulated expression of CD163 with a corresponding increase in PRRSV infection. These data indicate that the expression of CD163 on macrophages in different microenvironments, in vivo, may determine the replication efficiency and subsequent pathogenecity of PRRSV.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Macrófagos Alveolares/metabolismo , Síndrome Respiratória e Reprodutiva Suína/metabolismo , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Receptores de Superfície Celular/metabolismo , Suínos/metabolismo , Replicação Viral , Animais , Anticorpos Monoclonais/farmacologia , Linhagem Celular , Regulação da Expressão Gênica , Interleucina-10/farmacologia , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/virologia , Síndrome Respiratória e Reprodutiva Suína/virologia , Suínos/virologia , Acetato de Tetradecanoilforbol/farmacologia
11.
J Virol ; 82(19): 9306-17, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18632864

RESUMO

The inability to grow human noroviruses in cell culture has greatly impeded the studies of their pathogenesis and immunity. Vesiviruses, in the family Caliciviridae, grow efficiently in cell culture and encode a unique protein in the subgenomic region designated as leader of the capsid protein (LC). We hypothesized that LC might be associated with the efficient replication of vesiviruses in cell culture and promote the replication of human norovirus in cells. To test this hypothesis, a recombinant plasmid was engineered in which the LC region of feline calicivirus (FCV) was placed under the control of the cytomegalovirus promoter (pCI-LC) so that the LC protein could be provided in trans to replicating calicivirus genomes bearing a reporter gene. We constructed pNV-GFP, a recombinant plasmid containing a full-length NV genome with a green fluorescent protein (GFP) in the place of VP1. The transfection of pNV-GFP in MVA-T7-infected cells produced few GFP-positive cells detected by fluorescence microscopy and flow cytometry analysis. When pNV-GFP was cotransfected with pCI-LC in MVA-T7-infected cells, we observed an increase in the number of GFP-positive cells (ca. 3% of the whole-cell population). Using this cotransfection method with mutagenesis study, we identified potential cis-acting elements at the start of subgenomic RNA and the 3' end of NV genome for the virus replication. We conclude that LC may be a viral factor which promotes the replication of NV in cells, which could provide a clue to growing the fastidious human noroviruses in cell culture.


Assuntos
Calicivirus Felino/genética , Vírus Norwalk/metabolismo , Animais , Calicivirus Felino/metabolismo , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Técnicas Genéticas , Genoma Viral , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microscopia de Fluorescência/métodos , Modelos Genéticos , Plasmídeos/metabolismo , Proteínas Recombinantes/química , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA