Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
J Immunol ; 211(4): 601-611, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37395686

RESUMO

Retinoic acid (RA) is a fundamental vitamin A metabolite involved in regulating immune responses through the nuclear RA receptor (RAR) and retinoid X receptor. While performing experiments using THP-1 cells as a model for Mycobacterium tuberculosis infection, we observed that serum-supplemented cultures displayed high levels of baseline RAR activation in the presence of live, but not heat-killed, bacteria, suggesting that M. tuberculosis robustly induces the endogenous RAR pathway. Using in vitro and in vivo models, we have further explored the role of endogenous RAR activity in M. tuberculosis infection through pharmacological inhibition of RARs. We found that M. tuberculosis induces classical RA response element genes such as CD38 and DHRS3 in both THP-1 cells and human primary CD14+ monocytes via a RAR-dependent pathway. M. tuberculosis-stimulated RAR activation was observed with conditioned media and required nonproteinaceous factor(s) present in FBS. Importantly, RAR blockade by (4-[(E)-2-[5,5-dimethyl-8-(2-phenylethynyl)-6H-naphthalen-2-yl]ethenyl]benzoic acid), a specific pan-RAR inverse agonist, in a low-dose murine model of tuberculosis significantly reduced SIGLEC-F+CD64+CD11c+high alveolar macrophages in the lungs, which correlated with 2× reduction in tissue mycobacterial burden. These results suggest that the endogenous RAR activation axis contributes to M. tuberculosis infection both in vitro and in vivo and reveal an opportunity for further investigation of new antituberculosis therapies.


Assuntos
Mycobacterium tuberculosis , Receptores do Ácido Retinoico , Camundongos , Humanos , Animais , Receptores do Ácido Retinoico/metabolismo , Mycobacterium tuberculosis/metabolismo , Agonismo Inverso de Drogas , Tretinoína/farmacologia , Receptores X de Retinoides
2.
J Immunol ; 210(12): 1925-1937, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37098890

RESUMO

COVID-19 has accounted for more than 6 million deaths worldwide. Bacillus Calmette-Guérin (BCG), the existing tuberculosis vaccine, is known to induce heterologous effects over other infections due to trained immunity and has been proposed to be a potential strategy against SARS-CoV-2 infection. In this report, we constructed a recombinant BCG (rBCG) expressing domains of the SARS-CoV-2 nucleocapsid and spike proteins (termed rBCG-ChD6), recognized as major candidates for vaccine development. We investigated whether rBCG-ChD6 immunization followed by a boost with the recombinant nucleocapsid and spike chimera (rChimera), together with alum, provided protection against SARS-CoV-2 infection in K18-hACE2 mice. A single dose of rBCG-ChD6 boosted with rChimera associated with alum elicited the highest anti-Chimera total IgG and IgG2c Ab titers with neutralizing activity against SARS-CoV-2 Wuhan strain when compared with control groups. Importantly, following SARS-CoV-2 challenge, this vaccination regimen induced IFN-γ and IL-6 production in spleen cells and reduced viral load in the lungs. In addition, no viable virus was detected in mice immunized with rBCG-ChD6 boosted with rChimera, which was associated with decreased lung pathology when compared with BCG WT-rChimera/alum or rChimera/alum control groups. Overall, our study demonstrates the potential of a prime-boost immunization system based on an rBCG expressing a chimeric protein derived from SARS-CoV-2 to protect mice against viral challenge.


Assuntos
COVID-19 , Mycobacterium bovis , Animais , Camundongos , Vacina BCG/genética , Proteínas Recombinantes de Fusão/genética , SARS-CoV-2 , Vacinas Sintéticas , COVID-19/prevenção & controle , Mycobacterium bovis/genética
3.
Immunol Cell Biol ; 99(3): 309-322, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33068449

RESUMO

Respiratory syncytial virus (RSV) is the major cause of lower respiratory tract infections in children under 1 year. RSV vaccines are currently unavailable, and children suffering from multiple reinfections by the same viral strain fail to develop protective responses. Although RSV-specific antibodies can be detected upon infection, these have limited neutralizing capacity. Follicular helper T (Tfh) cells are specialized in providing signals to B cells and help the production and affinity maturation of antibodies, mainly via interleukin (IL) 21 secretion. In this study, we evaluated whether RSV could inhibit Tfh responses. We observed that Tfh cells fail to upregulate IL-21 production upon RSV infection. In the lungs, RSV infection downregulated the expression of IL-21/interleukin-21 receptor (IL-21R) in Tfh cells and upregulated programmed death-ligand 1 (PD-L1) expression in dendritic cells (DCs) and B cells. PD-L1 blockade during infection recovered IL-21R expression in Tfh cells and increased the secretion of IL-21 in a DC-dependent manner. IL-21 treatment decreased RSV viral load and lung inflammation, inducing the formation of tertiary lymphoid organs in the lung. It also decreased regulatory follicular T cells, and increased Tfh cells, B cells, antibody avidity and neutralization capacity, leading to an overall improved anti-RSV humoral response in infected mice. Passive immunization with purified immunoglobulin G from IL-21-treated RSV-infected mice protected against RSV infection. Our results unveil a pathway by which RSV affects Tfh cells by increasing PD-L1 expression on antigen-presenting cells, highlighting the importance of an IL-21-PD-L1 axis for the generation of protective responses to RSV infection.


Assuntos
Anticorpos Neutralizantes , Infecções por Vírus Respiratório Sincicial , Animais , Anticorpos Antivirais , Interleucinas , Camundongos , Infecções por Vírus Respiratório Sincicial/terapia , Células T Auxiliares Foliculares
4.
Nat Immunol ; 9(11): 1279-87, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18806793

RESUMO

Mice deficient in the interferon-gamma (IFN-gamma)-inducible, immunity-related GTPase Irgm1 have defective host resistance to a variety of intracellular pathogens. This greater susceptibility to infection is associated with impaired IFN-gamma-dependent macrophage microbicidal activity in vitro. Here we show that Irgm1 also regulated the survival of mature effector CD4(+) T lymphocytes by protecting them from IFN-gamma-induced autophagic cell death. Mice deficient in both IFN-gamma and Irgm1 were 'rescued' from the lymphocyte depletion and greater mortality that occurs in mice singly deficient in Irgm1 after mycobacterial infection. Our studies identify a feedback mechanism in the T helper type 1 response that limits the detrimental effects of IFN-gamma on effector T lymphocyte survival while promoting the antimicrobial functions of IFN-gamma.


Assuntos
Autofagia , Linfócitos T CD4-Positivos/imunologia , Proteínas de Ligação ao GTP/imunologia , Interferon gama/imunologia , Animais , Autofagia/genética , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/ultraestrutura , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Feminino , Proteínas de Ligação ao GTP/genética , Interferon gama/genética , Interferon gama/farmacologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium avium/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Tuberculose/imunologia
5.
J Immunol ; 200(4): 1434-1442, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29311364

RESUMO

IFN-stimulated gene 15 (ISG15) deficiency in humans leads to severe IFNopathies and mycobacterial disease, the latter being previously attributed to its extracellular cytokine-like activity. In this study, we demonstrate a novel role for secreted ISG15 as an IL-10 inducer, unique to primary human monocytes. A balanced ISG15-induced monocyte/IL-10 versus lymphoid/IFN-γ expression, correlating with p38 MAPK and PI3K signaling, was found using targeted in vitro and ex vivo systems analysis of human transcriptomic datasets. The specificity and MAPK/PI3K-dependence of ISG15-induced monocyte IL-10 production was confirmed in vitro using CRISPR/Cas9 knockout and pharmacological inhibitors. Moreover, this ISG15/IL-10 axis was amplified in leprosy but disrupted in human active tuberculosis (TB) patients. Importantly, ISG15 strongly correlated with inflammation and disease severity during active TB, suggesting its potential use as a biomarker, awaiting clinical validation. In conclusion, this study identifies a novel anti-inflammatory ISG15/IL-10 myeloid axis that is disrupted in active TB.


Assuntos
Citocinas/imunologia , Interleucina-10/imunologia , Leucócitos Mononucleares/imunologia , Tuberculose/imunologia , Ubiquitinas/imunologia , Humanos
6.
J Virol ; 92(10)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29514900

RESUMO

Dengue virus (DV) infection can cause either a self-limiting flu-like disease or a threatening hemorrhage that may evolve to shock and death. A variety of cell types, such as dendritic cells, monocytes, and B cells, can be infected by DV. However, despite the role of T lymphocytes in the control of DV replication, there remains a paucity of information on possible DV-T cell interactions during the disease course. In the present study, we have demonstrated that primary human naive CD4+ and CD8+ T cells are permissive for DV infection. Importantly, both T cell subtypes support viral replication and secrete viable virus particles. DV infection triggers the activation of both CD4+ and CD8+ T lymphocytes, but preactivation of T cells reduces the susceptibility of T cells to DV infection. Interestingly, the cytotoxicity-inducing protein granzyme A is highly secreted by human CD4+ but not CD8+ T cells after exposure to DV in vitro Additionally, using annexin V and polycaspase assays, we have demonstrated that T lymphocytes, in contrast to monocytes, are resistant to DV-induced apoptosis. Strikingly, both CD4+ and CD8+ T cells were found to be infected with DV in acutely infected dengue patients. Together, these results show that T cells are permissive for DV infection in vitro and in vivo, suggesting that this cell population may be a viral reservoir during the acute phase of the disease.IMPORTANCE Infection by dengue virus (DV) causes a flu-like disease that can evolve to severe hemorrhaging and death. T lymphocytes are important cells that regulate antibody secretion by B cells and trigger the death of infected cells. However, little is known about the direct interaction between DV and T lymphocytes. Here, we show that T lymphocytes from healthy donors are susceptible to infection by DV, leading to cell activation. Additionally, T cells seem to be resistant to DV-induced apoptosis, suggesting a potential role as a viral reservoir in humans. Finally, we show that both CD4+ and CD8+ T lymphocytes from acutely infected DV patients are infected by DV. Our results raise new questions about DV pathogenesis and vaccine development.


Assuntos
Apoptose/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/virologia , Vírus da Dengue/imunologia , Dengue/imunologia , Ativação Linfocitária/imunologia , Adolescente , Adulto , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Dengue/virologia , Vírus da Dengue/fisiologia , Feminino , Granzimas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Replicação Viral/imunologia , Adulto Jovem
7.
J Immunol ; 197(5): 1852-63, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27439514

RESUMO

The Toll-like and IL-1 family receptors play critical roles in innate and adaptive immunity against intracellular pathogens. Although previous data demonstrated the importance of TLRs and IL-1R signaling events for the establishment of an effective immune response to mycobacteria, the possible function of the adaptor molecule IL-1R-associated kinase (IRAK)-4 against this pathogen has not been addressed. In this study, we determined the role of IRAK-4 in signaling pathways responsible for controlling mycobacterial infections. This kinase is important for the production of IL-12 and TNF-α by macrophages and dendritic cells exposed to mycobacteria. Moreover, Mycobacterium bovis-infected IRAK-4-knockout macrophages displayed impaired MAPK and NF-κB activation. IL-1ß secretion and caspase-1 activation were also dependent on IRAK-4 signaling. Mice lacking IRAK-4 showed increased M. bovis burden in spleen, liver, and lungs and smaller liver granulomas during 60 d of infection compared with wild-type mice. Furthermore, 80% of IRAK-4(-/-) mice succumbed to virulent M. tuberculosis within 100 d following low-dose infection. This increased susceptibility to mycobacteria correlated with reduced IFN-γ/TNF-α recall responses by splenocytes, as well as fewer IL-12p70-producing APCs. Additionally, we observed that IRAK-4 is also important for the production of IFN-γ by CD4(+) T cells from infected mice. Finally, THP-1 cells treated with an IRAK-4 inhibitor and exposed to M. bovis showed reduced TNF-α and IL-12, suggesting that the results found in mice can be extended to humans. In summary, these data demonstrate that IRAK-4 is essential for innate and adaptive immunity and necessary for efficient control of mycobacterial infections.


Assuntos
Quinases Associadas a Receptores de Interleucina-1/deficiência , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Células Th1/patologia , Tuberculose/imunologia , Imunidade Adaptativa , Animais , Carga Bacteriana , Caspase 1/genética , Caspase 1/metabolismo , Linhagem Celular , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Humanos , Imunidade Inata , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Fígado/microbiologia , Fígado/patologia , Pulmão/microbiologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/imunologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/microbiologia , Mycobacterium bovis/crescimento & desenvolvimento , Mycobacterium bovis/imunologia , Mycobacterium bovis/patogenicidade , NF-kappa B/metabolismo , Transdução de Sinais , Baço/microbiologia , Células Th1/imunologia , Tuberculina/imunologia , Tuberculose/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Eur J Immunol ; 46(8): 1936-47, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27230303

RESUMO

Proinflammatory cytokines are critical mediators that control Mycobacterium tuberculosis (Mtb) growth during active tuberculosis (ATB). To further inhibit bacterial proliferation in diseased individuals, drug inhibitors of cell wall synthesis such as isoniazid (INH) are employed. However, whether INH presents an indirect effect on bacterial growth by regulating host cytokines during ATB is not well known. To examine this hypothesis, we used an in vitro human granuloma system generated with primary leukocytes from healthy donors adapted to model ATB. Intense Mtb proliferation in cell cultures was associated with monocyte/macrophage activation and secretion of IL-1ß and TNF. Treatment with INH significantly reduced Mtb survival, but altered neither T-cell-mediated Mtb killing, nor production of IL-1ß and TNF. However, blockade of both IL-1R1 and TNF signaling rescued INH-induced killing, suggesting synergistic roles of these cytokines in mediating control of Mtb proliferation. Additionally, mycobacterial killing by INH was highly dependent upon drug activation by the pathogen catalase-peroxidase KatG and involved a host PI3K-dependent pathway. Finally, experiments using coinfected (KatG-mutated and H37Rv strains) cells suggested that active INH does not directly enhance host-mediated killing of Mtb. Our results thus indicate that Mtb-stimulated host IL-1 and TNF have potential roles in TB chemotherapy.


Assuntos
Antituberculosos/farmacologia , Interleucina-1beta/imunologia , Isoniazida/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Receptores de Interleucina-1/imunologia , Fator de Necrose Tumoral alfa/imunologia , Proteínas de Bactérias/metabolismo , Células Cultivadas , Humanos , Macrófagos/metabolismo , Monócitos/metabolismo , Fosfatidilinositol 3-Quinases/imunologia , Receptores de Interleucina-1/antagonistas & inibidores , Tuberculose/tratamento farmacológico , Tuberculose/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores
9.
Front Immunol ; 15: 1331731, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38384473

RESUMO

The establishment of a virus infection is the result of the pathogen's ability to replicate in a hostile environment generated by the host's immune system. Here, we found that ISG15 restricts Dengue and Zika viruses' replication through the stabilization of its binding partner USP18. ISG15 expression was necessary to control DV replication driven by both autocrine and paracrine type one interferon (IFN-I) signaling. Moreover, USP18 competes with NS5-mediated STAT2 degradation, a major mechanism for establishment of flavivirus infection. Strikingly, reconstitution of USP18 in ISG15-deficient cells was sufficient to restore the STAT2's stability and restrict virus growth, suggesting that the IFNAR-mediated ISG15 activity is also antiviral. Our results add a novel layer of complexity in the virus/host interaction interface and suggest that NS5 has a narrow window of opportunity to degrade STAT2, therefore suppressing host's IFN-I mediated response and promoting virus replication.


Assuntos
Dengue , Interferon Tipo I , Infecção por Zika virus , Zika virus , Humanos , Interferon Tipo I/metabolismo , Infecção por Zika virus/genética , Replicação Viral , Dengue/genética , Ubiquitinas/metabolismo , Citocinas/metabolismo , Ubiquitina Tiolesterase/metabolismo , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo
10.
Nat Med ; 12(3): 330-4, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16415877

RESUMO

Control of inflammation is crucial to prevent damage to the host during infection. Lipoxins and aspirin-triggered lipoxins are crucial modulators of proinflammatory responses; however, their intracellular mechanisms have not been completely elucidated. We previously showed that lipoxin A4 (LXA4) controls migration of dendritic cells (DCs) and production of interleukin (IL)-12 in vivo. In the absence of LXA4 biosynthetic pathways, the resulting uncontrolled inflammation during infection is lethal, despite pathogen clearance. Here we show that lipoxins activate two receptors in DCs, AhR and LXAR, and that this activation triggers expression of suppressor of cytokine signaling (SOCS)-2. SOCS-2-deficient DCs are hyper-responsive to microbial stimuli, as well as refractory to the inhibitory actions of LXA4, but not to IL-10. Upon infection with an intracellular pathogen, SOCS-2-deficient mice had uncontrolled production of proinflammatory cytokines, decreased microbial proliferation, aberrant leukocyte infiltration and elevated mortality. We also show that SOCS-2 is a crucial intracellular mediator of the anti-inflammatory actions of aspirin-induced lipoxins in vivo.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Lipoxinas/farmacologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Encéfalo/citologia , Encéfalo/parasitologia , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Interleucina-12/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Hidrocarboneto Arílico/deficiência , Receptores de Hidrocarboneto Arílico/metabolismo , Baço/citologia , Baço/efeitos dos fármacos , Proteínas Supressoras da Sinalização de Citocina/deficiência , Proteínas Supressoras da Sinalização de Citocina/genética , Toxoplasma/patogenicidade
11.
Int J Biol Macromol ; 224: 1450-1459, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36441080

RESUMO

Ultrasensitive electroanalytical monitoring of interleukin-6 levels in serum samples has emerged as a valuable tool for the early diagnosis of inflammatory diseases. Despite its advantages, there is a lack of strategies for the label-free voltammetric determination of cytokines. Here, a novel chitosan/genipin modified fluorine tin oxide electrode was developed providing an in-situ hydrogel formation (FTO/CSG). This platform was applied for the detection of interleukin-6, a major pro-inflammatory cytokine. Transmission electron microscopy (TEM), cyclic voltammetry (CV), and electrochemical impedance spectroscopy (EIS) indicated genipin serves as an efficient green cross-linker to build the immunosensing platform (FTO/CSG/anti-IL-6). EIS showed an increase in charge transfer resistance from 326 to 1360 kΩ after the immobilization of anti-IL-6 antibodies. By square wave voltammetry, this method achieved a detection limit of 0.03 pg mL-1 with a wide linear range of 0.05-1000 pg mL-1. Additionally, it displayed a high selectivity index when tested in the presence of three inflammatory cytokines as interfering proteins: IL-12, IL-1ß, and TNF-α. The sample matrix effect showed a peak current variation lower than 5 %. The novel method was applied for the quantification of IL-6 in serum samples of septic mice. No statistical differences were observed between the standard ELISA and the proposed method using a confidence level of 95 %.


Assuntos
Técnicas Biossensoriais , Quitosana , Sepse , Animais , Camundongos , Interleucina-6 , Técnicas Eletroquímicas/métodos , Técnicas Biossensoriais/métodos , Biomarcadores , Eletrodos , Imunoensaio/métodos , Limite de Detecção
12.
Br J Pharmacol ; 180(11): 1460-1481, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36526272

RESUMO

BACKGROUND AND PURPOSE: Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Because pathogen-derived neuraminidase (NEU) stimulates neutrophils, we investigated whether host NEU can be targeted to regulate the neutrophil dysregulation observed in severe infections. EXPERIMENTAL APPROACH: The effects of NEU inhibitors on lipopolysaccharide (LPS)-stimulated neutrophils from healthy donors or COVID-19 patients were determined by evaluating the shedding of surface sialic acids, cell activation, and reactive oxygen species (ROS) production. Re-analysis of single-cell RNA sequencing of respiratory tract samples from COVID-19 patients also was carried out. The effects of oseltamivir on sepsis and betacoronavirus-induced acute lung injury were evaluated in murine models. KEY RESULTS: Oseltamivir and zanamivir constrained host NEU activity, surface sialic acid release, cell activation, and ROS production by LPS-activated human neutrophils. Mechanistically, LPS increased the interaction of NEU1 with matrix metalloproteinase 9 (MMP-9). Inhibition of MMP-9 prevented LPS-induced NEU activity and neutrophil response. In vivo, treatment with oseltamivir fine-tuned neutrophil migration and improved infection control as well as host survival in peritonitis and pneumonia sepsis. NEU1 also is highly expressed in neutrophils from COVID-19 patients, and treatment of whole-blood samples from these patients with either oseltamivir or zanamivir reduced neutrophil overactivation. Oseltamivir treatment of intranasally infected mice with the mouse hepatitis coronavirus 3 (MHV-3) decreased lung neutrophil infiltration, viral load, and tissue damage. CONCLUSION AND IMPLICATIONS: These findings suggest that interplay of NEU1-MMP-9 induces neutrophil overactivation. In vivo, NEU may serve as a host-directed target to dampen neutrophil dysfunction during severe infections.


Assuntos
COVID-19 , Sepse , Humanos , Camundongos , Animais , Oseltamivir/efeitos adversos , Zanamivir/efeitos adversos , Neuraminidase/metabolismo , Neuraminidase/farmacologia , Neutrófilos , Metaloproteinase 9 da Matriz/metabolismo , Espécies Reativas de Oxigênio , Lipopolissacarídeos/farmacologia , Sepse/induzido quimicamente
13.
Antimicrob Agents Chemother ; 56(5): 2259-67, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22330919

RESUMO

Nanoenabled drug delivery systems against tuberculosis (TB) are thought to control pathogen replication by targeting antibiotics to infected tissues and phagocytes. However, whether nanoparticle (NP)-based carriers directly interact with Mycobacterium tuberculosis and how such drug delivery systems induce intracellular bacterial killing by macrophages is not defined. In the present study, we demonstrated that a highly hydrophobic citral-derived isoniazid analogue, termed JVA, significantly increases nanoencapsulation and inhibits M. tuberculosis growth by enhancing intracellular drug bioavailability. Importantly, confocal and atomic force microscopy analyses revealed that JVA-NPs associate with both intracellular M. tuberculosis and cell-free bacteria, indicating that NPs directly interact with the bacterium. Taken together, these data reveal a nanotechnology-based strategy that promotes antibiotic targeting into replicating extra- and intracellular mycobacteria, which could actively enhance chemotherapy during active TB.


Assuntos
Antituberculosos/farmacologia , Isoniazida/análogos & derivados , Isoniazida/farmacologia , Macrófagos/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Nanopartículas , Animais , Disponibilidade Biológica , Células Cultivadas , Composição de Medicamentos , Sistemas de Liberação de Medicamentos/métodos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ácido Láctico/química , Macrófagos/microbiologia , Camundongos , Microscopia de Força Atômica , Microscopia Confocal , Mycobacterium tuberculosis/fisiologia , Tamanho da Partícula , Fagocitose , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Tuberculose Pulmonar/tratamento farmacológico , Tuberculose Pulmonar/microbiologia
14.
J Virol ; 85(11): 5374-83, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21450836

RESUMO

A recent (2007 to 2009) dengue outbreak caused by dengue virus (DENV) in Paraguay presented unusual severe clinical outcomes associated with 50% mortality rates. Although it has been reported that inflammatory responses influence the severity of dengue virus infection (T. Pang, M. J. Cardosa, and M. G. Guzman, Immunol. Cell Biol. 85:43-45, 2007), there remains a paucity of information on virus-innate immunity interactions influencing clinical outcome. Using human dendritic cells from a major innate immune cell population as an in vitro model, we have investigated signature cytokine responses as well as infectivity-replicative profiles of DENV clinical isolates from either a nonfatal case of classical dengue fever (strain DENV3/290; isolated in Brazil in 2002) or a fatal case of dengue fever with visceral complications isolated in Paraguay in 2007 (strain DENV3/5532). Strain DENV3/5532 was found to display significantly higher replicative ability than DENV3/290 in monocyte-derived dendritic cells (mdDCs). In addition, compared to DENV3/290 results, mdDCs exposed to DENV3/5532 showed increased production of proinflammatory cytokines associated with higher rates of programmed cell death, as shown by annexin V staining. The observed phenotype was due to viral replication, and tumor necrosis factor alpha (TNF-α) appears to exert a protective effect on virus-induced mdDC apoptosis. These results suggest that the DENV3/5532 strain isolated from the fatal case replicates within human dendritic cells, modulating cell survival and synthesis of inflammatory mediators.


Assuntos
Apoptose , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/virologia , Vírus da Dengue/patogenicidade , Dengue/virologia , Brasil , Vírus da Dengue/isolamento & purificação , Humanos , Dados de Sequência Molecular , Paraguai , RNA Viral/genética , Análise de Sequência de DNA , Replicação Viral
15.
Neuroimmunomodulation ; 19(6): 377-85, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23075771

RESUMO

BACKGROUND: Cytokines have been shown to be involved in traumatic brain injury (TBI). We investigated the independent association between serum levels of IL-10 and TNF-α and hospital mortality of patients with severe TBI. METHODS: Serum IL-10 and TNF-α levels were determined after a median period (interquartile range (IQ) 25-75) of 10 h (IQ 5-18) after severe TBI in 93 consecutive patients and in randomly selected patients with mild (n = 18) and moderate (n = 16) TBI. In patients with severe TBI, additional blood samples were analyzed 30 h (IQ 22-37) and 68 h (IQ 55-78) after TBI. Age, gender, computed tomography findings, Glasgow Coma Scale score (GCS) and pupil reactions at admission, associated trauma and hospital mortality were collected. RESULTS: Elevated serum levels of IL-10, but not TNF-α, correlated significantly with GCS severity (R(2) coefficient, p < 0.0001) and were found to be associated with hospital mortality in patients with severe TBI. Elevated IL-10 remained associated with mortality (p = 0.01) in a subset of patients with isolated severe TBI (n = 74). Multiple logistic regression analysis showed that higher IL-10 levels (>90 pg/ml) at 10 or 30 h after TBI were 6 times (odds ratio (OR) 6.2, 95% confidence interval (CI) 1.2-25.1, p = 0.03) and 5 times (OR 5.4, 95% CI 1.2-25.1, p = 0.03), respectively, more frequently associated with hospital mortality than lower levels (<50 pg/ml), independently of age, GCS as well as pupil reactions at admission and associated trauma. CONCLUSIONS: Serum IL-10 levels may be a useful marker for severe TBI prognosis.


Assuntos
Lesões Encefálicas/diagnóstico , Lesões Encefálicas/imunologia , Escala de Coma de Glasgow , Escala de Gravidade do Ferimento , Interleucina-10/biossíntese , Regulação para Cima/imunologia , Adolescente , Adulto , Idoso , Biomarcadores/sangue , Lesões Encefálicas/sangue , Feminino , Humanos , Interleucina-10/sangue , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Adulto Jovem
16.
J Infect Dis ; 204(3): 478-86, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21742848

RESUMO

BACKGROUND: Chagas disease is caused by the protozoan parasite Trypanosoma cruzi, which exhibits a high genetic variability. TcI, TcII, or mixed TcI/TcII strains may be found during acute human infection while mainly TcII parasites are present at the chronic stage of disease. In a previously studied Chagas disease outbreak, we identified mixed TcI/TcII strains in the vector Triatoma tibiamaculata and only TcII strains in infected humans, indicating that T. cruzi populations may be selected within the human host. METHODS: Utilizing molecular typing and cell biology techniques, we investigated the interaction of TcI, TcII, and mixed TcI/TcII strains with macrophages, an important cell population implicated in controlling protozoan infection. RESULTS: TcII but not TcI strains were selected by both human and murine macrophages in vitro and by peritoneal cavity cells in vivo. Biological analysis revealed that, compared with TcI, TcII strains display higher infective and multiplicative ability as well as lower doubling time inside macrophages. However, TcI and TcII strains present similar susceptibility to interferon-γ-activated macrophages in vitro. CONCLUSIONS: Taken together, our results reveal the existence of an intracellular selection process in macrophages that favors TcII, but not TcI, when infection occurs with vector-derived mixed TcI/TcII strains.


Assuntos
Doença de Chagas/parasitologia , Macrófagos/parasitologia , Trypanosoma cruzi/classificação , Doença Aguda , Animais , Anticorpos Antiprotozoários/imunologia , Humanos , Interferon gama/farmacologia , Ativação de Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Filogenia
17.
Commun Biol ; 5(1): 626, 2022 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-35752645

RESUMO

Given the discontinuation of various first-line drugs for visceral leishmaniasis (VL), large-scale in vivo drug screening, establishment of a relapse model in rodents, immunophenotyping, and transcriptomics were combined to study persistent infections and therapeutic failure. Double bioluminescent/fluorescent Leishmania infantum and L. donovani reporter lines enabled the identification of long-term hematopoietic stem cells (LT-HSC) as a niche in the bone marrow with remarkably high parasite burdens, a feature confirmed for human hematopoietic stem cells (hHSPC). LT-HSC are more tolerant to antileishmanial drug action and serve as source of relapse. A unique transcriptional 'StemLeish' signature in these cells was defined by upregulated TNF/NF-κB and RGS1/TGF-ß/SMAD/SKIL signaling, and a downregulated oxidative burst. Cross-species analyses demonstrated significant overlap with human VL and HIV co-infected blood transcriptomes. In summary, the identification of LT-HSC as a drug- and oxidative stress-resistant niche, undergoing a conserved transcriptional reprogramming underlying Leishmania persistence and treatment failure, may open therapeutic avenues for leishmaniasis.


Assuntos
Leishmaniose Visceral , Parasitos , Animais , Células-Tronco Hematopoéticas , Leishmaniose Visceral/tratamento farmacológico , Leishmaniose Visceral/parasitologia , Recidiva , Falha de Tratamento
18.
Front Immunol ; 13: 1042463, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36311766

RESUMO

Zika virus (ZIKV) is a single-strand RNA mosquito-borne flavivirus with significant public health impact. ZIKV infection induces double-strand DNA breaks (DSBs) in human neural progenitor cells that may contribute to severe neuronal manifestations in newborns. The DNA-PK complex plays a critical role in repairing DSBs and in the innate immune response to infection. It is unknown, however, whether DNA-PK regulates ZIKV infection. Here we investigated the role of DNA-PKcs, the catalytic subunit of DNA-PK, during ZIKV infection. We demonstrate that DNA-PKcs restricts the spread of ZIKV infection in human epithelial cells. Increased ZIKV replication and spread in DNA-PKcs deficient cells is related to a notable decrease in transcription of type I and III interferons as well as IFIT1, IFIT2, and IL6. This was shown to be independent of IRF1, IRF3, or p65, canonical transcription factors necessary for activation of both type I and III interferon promoters. The mechanism of DNA-PKcs to restrict ZIKV infection is independent of DSB. Thus, these data suggest a non-canonical role for DNA-PK during Zika virus infection, acting downstream of IFNs transcription factors for an efficient antiviral immune response.


Assuntos
Infecção por Zika virus , Zika virus , Recém-Nascido , Animais , Humanos , Zika virus/fisiologia , Replicação Viral , Interferons/farmacologia , Antivirais/uso terapêutico , DNA
19.
bioRxiv ; 2022 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-33200130

RESUMO

Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Neuraminidase (NEU)-mediated cleavage of surface sialic acid has been demonstrated to regulate leukocyte responses. Here, we report that antiviral NEU inhibitors constrain host NEU activity, surface sialic acid release, ROS production, and NETs released by microbial-activated human neutrophils. In vivo, treatment with Oseltamivir results in infection control and host survival in peritonitis and pneumonia models of sepsis. Single-cell RNA sequencing re-analysis of publicly data sets of respiratory tract samples from critical COVID-19 patients revealed an overexpression of NEU1 in infiltrated neutrophils. Moreover, Oseltamivir or Zanamivir treatment of whole blood cells from severe COVID-19 patients reduces host NEU-mediated shedding of cell surface sialic acid and neutrophil overactivation. These findings suggest that neuraminidase inhibitors can serve as host-directed interventions to dampen neutrophil dysfunction in severe infections.

20.
J Exp Med ; 202(12): 1715-24, 2005 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-16365150

RESUMO

To investigate the role of Toll-like receptor (TLR)9 in the immune response to mycobacteria as well as its cooperation with TLR2, a receptor known to be triggered by several major mycobacterial ligands, we analyzed the resistance of TLR9(-/-) as well as TLR2/9 double knockout mice to aerosol infection with Mycobacterium tuberculosis. Infected TLR9(-/-) but not TLR2(-/-) mice displayed defective mycobacteria-induced interleukin (IL)-12p40 and interferon (IFN)-gamma responses in vivo, but in common with TLR2(-/-) animals, the TLR9(-/-) mice exhibited only minor reductions in acute resistance to low dose pathogen challenge. When compared with either of the single TLR-deficient animals, TLR2/9(-/-) mice displayed markedly enhanced susceptibility to infection in association with combined defects in proinflammatory cytokine production in vitro, IFN-gamma recall responses ex vivo, and altered pulmonary pathology. Cooperation between TLR9 and TLR2 was also evident at the level of the in vitro response to live M. tuberculosis, where dendritic cells and macrophages from TLR2/9(-/-) mice exhibited a greater defect in IL-12 response than the equivalent cell populations from single TLR9-deficient animals. These findings reveal a previously unappreciated role for TLR9 in the host response to M. tuberculosis and illustrate TLR collaboration in host resistance to a major human pathogen.


Assuntos
Imunidade Inata/imunologia , Mycobacterium tuberculosis/imunologia , Células Th1/imunologia , Receptor Toll-Like 9/imunologia , Tuberculose/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Citocinas/metabolismo , Primers do DNA , Citometria de Fluxo , Pulmão/metabolismo , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor Toll-Like 9/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa