Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 744
Filtrar
1.
Vet Microbiol ; 296: 110166, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38968694

RESUMO

Streptococcus suis (S. suis) disease is a prevalent zoonotic infectious threat that elicits a systemic inflammatory response in both swine and humans, frequently culminating in high mortality rates. The excessive inflammation triggered by S. suis infection can precipitate tissue damage and sudden death; however, a comprehensive strategy to mitigate this inflammatory response remains elusive. Our study examines the role of NLRP6 in S. suis infection, with a particular focus on its involvement in pathogen regulation. A marked upregulation of NLRP6 was observed in peritoneal macrophages post-infection with S. suis SC19 strain, consequently activating the NLRP6 inflammasome. Furthermore, SC19 infection was found to augment the secretion of pro-inflammatory cytokines IL-1ß via NLRP6 activation, while NLRP6 deficiency mitigates the invasion and adhesion of SC19 to macrophages. In vivo models revealed that NLRP6 deletion enhanced survival rates of SC19-infected mice, alongside a reduction in tissue bacterial load and inflammatory cytokine levels. NLRP6-/- mice were shown to exhibit attenuated inflammatory responses in pulmonary, hepatic, and splenic tissues post-SC19 infection, as evidenced by lower inflammation scores. Flow cytometry analyses further substantiated that NLRP6 is involved in modulating macrophage and neutrophil recruitment during infection. Our findings suggest that NLRP6 negatively regulates host resistance against S. suis infection; its absence results in reduced mortality, bacterial colonization, and a milder inflammatory response. Elucidating the mechanism of NLRP6 in S. suis-induced inflammation provides novel insights and theoretical underpinnings for the prophylaxis and therapeutics of S. suis diseases.


Assuntos
Camundongos Endogâmicos C57BL , Infecções Estreptocócicas , Streptococcus suis , Streptococcus suis/imunologia , Streptococcus suis/patogenicidade , Streptococcus suis/genética , Animais , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Camundongos , Camundongos Knockout , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Inflamassomos/imunologia , Inflamassomos/genética , Citocinas/metabolismo , Citocinas/genética , Inflamação/imunologia , Feminino , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Receptores de Superfície Celular
2.
mSystems ; 9(7): e0048424, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38934644

RESUMO

Corynebacterium pseudotuberculosis (C. p), a facultative intracellular bacterium, is an important zoonotic pathogen that causes abscesses and pyogenic granulomas. The relationship between gut microbiota and host health or diseases has received increasing attention. However, the role of gut microbiota in the process of C. p infection is still unclear. In this study, we established a C. p infection model in C57BL/6 mice and examined the impact of preemptive oral administration Lactobacillus acidophilus (L. acidophilus) on infection. Our findings revealed that C. p infection led to pronounced pathological alterations in the liver and kidneys, characterized by abscess formation, intense inflammatory responses, and bacterial overload. Remarkably, these deleterious effects were greatly relieved by oral administration of L. acidophilus before infection with C. p. Additionally, we further found that during C. p infection, peritoneal macrophages (PMs) of mice orally administered with L. acidophilus accumulated more rapidly at sites of infection. Furthermore, our results showed that PMs from mice with oral L. acidophilus administration showed a stronger C. p clearance effect, and this was mediated by high expression of LC3-II protein. Meanwhile, oral administration of L. acidophilus protected the gut microbiota disorder in C57BL/6 mice caused by C. p infection. In summary, our study demonstrates that oral administration of L. acidophilus confers effective protection against C. p infection in C57BL/6 mice by modulating macrophage autophagy, thereby augmenting bacterial clearance and preserving gut microbiota and function stability. These findings position L. acidophilus as a viable probiotic candidate for the clinical prevention of C. p infection. IMPORTANCE: Corynebacterium pseudotuberculosis (C. p) is known to induce a range of chronic diseases in both animals and humans. Currently, clinical treatment for C. p infection mainly relies on antibiotic therapy or surgical intervention. However, excessive use of antibiotics may increase the risk of drug-resistant strains, and the effectiveness of treatment remains unsatisfactory. Furthermore, surgical procedures do not completely eradicate pathogens and can easily cause environmental pollution. Probiotic interventions are receiving increasing attention for improving the body's immune system and maintaining health. In this study, we established a C. p infection model in C57BL/6 mice to explore the impact of Lactobacillus acidophilus during C. p infection. Our results showed that L. acidophilus effectively protected against C. p infection by regulating the autophagy of macrophages and maintaining intestinal microbiota homeostasis. This study may provide a new strategy for the prevention of C. p infection.


Assuntos
Autofagia , Infecções por Corynebacterium , Corynebacterium pseudotuberculosis , Microbioma Gastrointestinal , Lactobacillus acidophilus , Camundongos Endogâmicos C57BL , Animais , Autofagia/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Lactobacillus acidophilus/fisiologia , Camundongos , Infecções por Corynebacterium/prevenção & controle , Infecções por Corynebacterium/microbiologia , Homeostase/efeitos dos fármacos , Probióticos/administração & dosagem , Probióticos/farmacologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Modelos Animais de Doenças
3.
Pancreas ; 53(7): e617-e626, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38696351

RESUMO

BACKGROUND: Although the involvement of intestinal microbiota in innate immunity has been reported recently, the pathogenicity of autoimmune pancreatitis (AIP) remains unclear. This study aimed to investigate whether probiotics ameliorate inflammation in AIP through interactions with innate immunity. MATERIALS AND METHODS: The AIP mouse model was generated by intraperitoneal administration of Escherichia coli to C56BL/6 female mice. Alterations in the intestinal microbiota in the AIP group were evaluated using high-throughput sequencing. Peritoneal macrophages (PMs) were collected and cocultured in vitro with Lactobacillus gasseri (LG) or ligands of Toll-like receptors (TLRs). LG was administered intraperitoneally to AIP model mice, and pancreatitis activity was evaluated to examine the ameliorative effects of LG. RESULTS: In the AIP model mice, inflammation was significantly induced in the pancreas, and the intestinal microbiota was altered with decreased LG. Antimicrobial treatment suppressed pancreatitis. In vitro, E. coli stimulation increased inflammatory cytokine expression, which was significantly decreased when the LG or TLR7 ligand was cocultured with PMs. Intraperitoneal administration of LG to AIP model mice significantly suppressed pancreatitis. CONCLUSION: The mouse model demonstrated the involvement of intestinal microbiota in pancreatitis, and LG administration suppressed pancreatitis, possibly through TLR7 signaling in PMs. LG may be a helpful probiotic for treating AIP.


Assuntos
Pancreatite Autoimune , Modelos Animais de Doenças , Microbioma Gastrointestinal , Imunidade Inata , Macrófagos Peritoneais , Camundongos Endogâmicos C57BL , Probióticos , Animais , Microbioma Gastrointestinal/imunologia , Probióticos/administração & dosagem , Probióticos/farmacologia , Feminino , Pancreatite Autoimune/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Citocinas/metabolismo , Camundongos , Escherichia coli/imunologia , Receptor 7 Toll-Like/imunologia , Pâncreas/imunologia , Pâncreas/patologia , Lactobacillus/imunologia
4.
Microb Pathog ; 183: 106294, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37567327

RESUMO

The CXCL8/CXCR1 axis in conjoint with the free radicals and anti-oxidants dictates the severity of inflammation caused by the bacteria, Staphylococcus aureus. S.aureus mediated inflammatory processes is regulated by NF-κB and its product, iNOS. The objective of this study was to examine the effects of inhibition of NF-κB and iNOS on CXCL8/CXCR1, alteration in M1/M2 polarization of macrophages and associated inflammatory responses during S.aureus infection in vitro. For this, the murine peritoneal macrophages were pretreated with NF-κB inhibitor, Pyrrolidine dithiocarbamate (PDTC) and iNOS inhibitor, L-N-monomethyl arginine (LNMMA), either alone or in combination, followed by time-dependent S.aureus infection. The chemotactic migrations of macrophages were determined by the agarose spot assay. The iNOS, NF-κB and CXCR1 protein expressions were evaluated. The ROS level (superoxide, H2O2, NO) and antioxidant activities (SOD, CAT, GSH, arginase) were measured. The intra-macrophage phagoctyic activity had been analyzed by confocal microscopy. S.aureus activated macrophages showed increased iNOS expression that symbolizes M1 characterization of macrophages. The results suggest that the combination treatment of LNMMA + PDTC was effective in diminution of CXCL8 production and CXCR1 expression through downregulation of NF-κB and iNOS signaling pathway. Consequently, there was decrement in macrophage migration, reduced ROS generation, elevated antioxidant enzyme activity as well as bacterial phagocytosis at 90 min post bacterial infection. The increased arginase activity further proves the switch from pro-inflammatory M1 to anti-inflammatory M2 polarization of macrophages. Concludingly, the combination of PDTC + LNMMA could resolve S.aureus mediated inflammation through mitigation of CXCL8/CXCR1 pathway switching from M1 to M2 polarization.


Assuntos
Macrófagos Peritoneais , Infecções Estafilocócicas , Camundongos , Animais , Macrófagos Peritoneais/microbiologia , Staphylococcus aureus/metabolismo , ômega-N-Metilarginina/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Espécies Reativas de Oxigênio/metabolismo , NF-kappa B/metabolismo , Peróxido de Hidrogênio/metabolismo , Arginase/metabolismo , Citocinas/metabolismo , Infecções Estafilocócicas/microbiologia , Receptores de Interleucina-8A/metabolismo , Inflamação/metabolismo
5.
Cell Rep ; 38(8): 110414, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35196496

RESUMO

Inflammasome activation exacerbates infectious disease caused by pathogens such as Listeria monocytogenes, Staphylococcus aureus, and severe acute respiratory syndrome coronavirus 2. Although these pathogens activate host inflammasomes to regulate pathogen expansion, the mechanisms by which pathogen toxins contribute to inflammasome activation remain poorly understood. Here we show that activation of inflammasomes by Listeria infection is promoted by amino acid residue T223 of listeriolysin O (LLO) independently of its pore-forming activity. LLO T223 is critical for phosphorylation of the inflammasome adaptor ASC at amino acid residue Y144 through Lyn-Syk signaling, which is essential for ASC oligomerization. Notably, a Listeria mutant expressing LLO T223A is impaired in inducing ASC phosphorylation and inflammasome activation. Furthermore, the virulence of LLO T223A mutant is markedly attenuated in vivo due to impaired ability to activate the inflammasome. Our results reveal a function of a pathogen toxin that exacerbates infection by promoting phosphorylation of ASC.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Inflamassomos/metabolismo , Listeria monocytogenes/patogenicidade , Transdução de Sinais , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Proteínas Adaptadoras de Sinalização CARD/química , Proteínas Adaptadoras de Sinalização CARD/deficiência , Proteínas Adaptadoras de Sinalização CARD/genética , Edição de Genes , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Proteínas Hemolisinas/química , Proteínas Hemolisinas/genética , Interleucina-18/metabolismo , Listeria monocytogenes/metabolismo , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Sítio-Dirigida , Fosforilação , Quinase Syk/genética , Quinase Syk/metabolismo , Virulência , Quinases da Família src/genética , Quinases da Família src/metabolismo
6.
Cell Immunol ; 372: 104483, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35085880

RESUMO

The occurring in SR-A/CD204- or CD36-deficient mice increased susceptibility to infections with Staphylococcus aureus (Sa) had traditionally been ascribed to the impairment of macrophage-mediated phagocytosis, which is, however, inconsistent with low effectiveness of unopsonized Sa killing within macrophages and redundant roles of both receptors in this process. We have found that Sa-stimulated cytokine production in mouse macrophages seems to be exclusively mediated by TLR2, mainly from within endosomes in response to Sa-derived lipoteichoic acid. By driving endocytic trafficking of TLR2 and its ligands through the clathrin-dependent pathway, CD36 and SR-A sensitize macrophages to activation by Sa as well as regulate the type and amount of cytokines produced. Additionally, upon direct Sa binding, both receptors autonomously generate anti-inflammatory signaling. Consequently, the delayed induction of acute inflammation in knockout mice may allow for the initial, uncontrolled multiplication of bacteria, stimulating excessive, septic shock-inducing production of inflammatory cytokines in later stages of infection.


Assuntos
Antígenos CD36/imunologia , Citocinas/biossíntese , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Receptores Depuradores Classe A/imunologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Animais , Antígenos CD36/deficiência , Antígenos CD36/genética , Endocitose/imunologia , Ligantes , Receptores de Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Reconhecimento de Padrão/imunologia , Receptores Depuradores Classe A/deficiência , Receptores Depuradores Classe A/genética , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/imunologia
7.
Chem Biol Interact ; 351: 109713, 2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-34699765

RESUMO

Leishmaniasis is an infectious-parasitic disease caused by the protozoan Leishmania spp. The available treatments are based upon expensive drugs bearing adverse side-effects. The search for new therapeutic alternatives that present a more effective action without causing adverse effects to the patient is therefore important. The objective of this study was to evaluate the in vitro effect of botryosphaeran, a (1 â†’ 3)(1 â†’ 6)-ß-D-glucan, on the promastigote and intracellular amastigote forms of Leishmania amazonensis. The direct activity of botryosphaeran on promastigote forms was evaluated in vitro and inhibited proliferation, the IC50 7 µg/mL in 48 h was calculated. After 48 h treatment, botryosphaeran induced nitric oxide production (NO), caused mitochondrial membrane hyperpolarization, increased reactive oxygen species (ROS), and accumulation of lipid vesicles in promastigotes, resulting in apoptosis, necrosis and autophagy, and was accompanied by morphological and ultrastructural changes. The range of concentrations used did not alter the viability of peritoneal macrophages from BALB/c mice and erythrocytes of sheep. Botryosphaeran was able to reduce the number of infected macrophages and the number of amastigotes per macrophage at 12.5 µg/mL (50.75% ± 6.48), 25 µg/mL (55.66% ± 3.93) and 50 µg/mL (72.9% ± 6.98), and IC50 9.3 µg/mL (±0.66) for intracellular amastigotes forms. The leishmanicidal effect was due to activation of NF-κB and promoted an increase in pro-inflammatory cytokines (TNF-α and IL-6), iNOS and microbial-derived ROS and NO, in addition to decreasing the levels of SOD. Based upon the data obtained, we infer that botryosphaeran exerted an active leishmanicidal and immunomodulatory effect, acting on promastigotes through autophagic, apoptotic and necrosis processes, and in the intracellular amastigote form, through the action of ROS and NO.


Assuntos
Apoptose/efeitos dos fármacos , Glucanos/farmacologia , Fatores Imunológicos/farmacologia , Leishmania/efeitos dos fármacos , NF-kappa B/metabolismo , Tripanossomicidas/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Interleucina-6/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos Endogâmicos BALB C , Necrose/induzido quimicamente , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Testes de Sensibilidade Parasitária , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Dev Comp Immunol ; 127: 104292, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34656643

RESUMO

Vibrio harveyi is a zoonotic pathogen that can infect humans through wounds and cause severe inflammatory responses. Previous studies have reported that the Toll like receptors (TLR) mediated MAPK, AKT and NF-κB signaling pathways are involved in innate immune system resistance to pathogen invasion. However, the molecular mechanism of these pathways, as well as their involvement in V. harveyi infection remains elusive. This study established a V. harveyi infection model using murine peritoneal macrophages (PMs). Various techniques, including western blotting, ELISA, RT-qPCR, immunofluorescence, inhibition assays, were used to explore the roles of TLRs, MAPK, AKT and NF-κB signaling pathways in V. harveyi-induced inflammatory responses. ELISA assays showed that V. harveyi infection triggered proinflammatory cytokines secretion in PMs. RT-qPCR and inhibition assays showed that TLR2 participated in V. harveyi infection and up-regulated the proinflammatory cytokines secretion in murine PMs. Western blotting data showed that the phosphorylation of p38, JNK, AKT, and NF-κB p65 were significantly increased partly mediated by TLR2. In addition, immunofluorescence assays revealed that the NF-κB p65 translocated into nucleus in response to V. harveyi infection. The secretion of IL-1ß, IL-6, IL-12, and TNF-α were considerably reduced when the p38 MAPK and NF-κB signaling pathways were blocked, whereas blocking of AKT significantly increased the expression of IL-1ß, IL-6, IL-12, and TNF-α. These findings indicate that V. harveyi infection induces inflammatory responses in murine PMs via activation of p38 MAPK and NF-κB pathways, which are partly mediated by TLR2, but are inhibited by PI3K/AKT pathways.


Assuntos
Citocinas , Macrófagos Peritoneais , NF-kappa B , Vibrioses , Proteínas Quinases p38 Ativadas por Mitógeno , Animais , Citocinas/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Camundongos , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Vibrio , Vibrioses/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Acta Biochim Biophys Sin (Shanghai) ; 53(12): 1590-1601, 2021 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-34569606

RESUMO

Vibrio harveyi, an important zoonotic pathogen, can infect wounds and cause inflammatory response. Understanding the inflammatory response pathways could facilitate the exploration of molecular mechanisms for treating V. harveyi infection. NLR family pyrin domain-containing 3 (NLRP3) inflammasome is involved in the interaction between hosts and pathogenic microorganisms and could be sensed by various pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). Nonetheless, the function of NLRP3 inflammasome in V. harveyi infection remains unclear. In the present study, we established a V. harveyi infection model using murine peritoneal macrophages (PMs). Various techniques, including western blot analysis, enzyme-linked immunosorbent assay (ELISA), RT-qPCR, immunofluorescence, and inhibition assays, were used to explore the molecular mechanism of V. harveyi-induced inflammation. The results showed that many inflammatory cytokines participated in V. harveyi infection, with interleukin (IL)-1ß being the most abundant. Pan-caspase inhibitor pretreatment significantly decreased the secretion of IL-1ß in murine PMs. Moreover, the identification of V. harveyi involved a large number of NLR molecules, especially the NLRP3 receptor, and further studies revealed that NLPR3 inflammasome was activated by V. harveyi infection, as evidenced by puncta-like NLRP3 surrounding cell nuclear, ASC specks in the nucleus and cytoplasm, and ASC oligomerization. Inhibition of NLRP3 inflammasome impaired the release of mature IL-1ß in V. harveyi-infected murine PMs. Furthermore, blocking the secretion of mature IL-1ß could markedly decrease the release of other proinflammatory cytokines, including IL-6, IL-12, and tumor necrosis factor-α. Overall, these data indicated that NLRP3 inflammasome was activated in response to V. harveyi infection and enhanced inflammatory response by promoting IL-1ß secretion in murine PMs.


Assuntos
Infecções por Bactérias Gram-Negativas/metabolismo , Inflamação/metabolismo , Inflamação/microbiologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Vibrio/patogenicidade , Animais , Caspase 1/metabolismo , Células Cultivadas , Citocinas/metabolismo , Feminino , Interleucina-1beta/metabolismo , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/agonistas , Transdução de Sinais , Fatores de Tempo
10.
Front Immunol ; 12: 688257, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34497601

RESUMO

We present a stochastic mathematical model of the intracellular infection dynamics of Bacillus anthracis in macrophages. Following inhalation of B. anthracis spores, these are ingested by alveolar phagocytes. Ingested spores then begin to germinate and divide intracellularly. This can lead to the eventual death of the host cell and the extracellular release of bacterial progeny. Some macrophages successfully eliminate the intracellular bacteria and will recover. Here, a stochastic birth-and-death process with catastrophe is proposed, which includes the mechanism of spore germination and maturation of B. anthracis. The resulting model is used to explore the potential for heterogeneity in the spore germination rate, with the consideration of two extreme cases for the rate distribution: continuous Gaussian and discrete Bernoulli. We make use of approximate Bayesian computation to calibrate our model using experimental measurements from in vitro infection of murine peritoneal macrophages with spores of the Sterne 34F2 strain of B. anthracis. The calibrated stochastic model allows us to compute the probability of rupture, mean time to rupture, and rupture size distribution, of a macrophage that has been infected with one spore. We also obtain the mean spore and bacterial loads over time for a population of cells, each assumed to be initially infected with a single spore. Our results support the existence of significant heterogeneity in the germination rate, with a subset of spores expected to germinate much later than the majority. Furthermore, in agreement with experimental evidence, our results suggest that most of the spores taken up by macrophages are likely to be eliminated by the host cell, but a few germinated spores may survive phagocytosis and lead to the death of the infected cell. Finally, we discuss how this stochastic modelling approach, together with dose-response data, allows us to quantify and predict individual infection risk following exposure.


Assuntos
Antraz/microbiologia , Bacillus anthracis/patogenicidade , Macrófagos Peritoneais/microbiologia , Modelos Biológicos , Esporos Bacterianos/patogenicidade , Animais , Antraz/imunologia , Antraz/patologia , Bacillus anthracis/crescimento & desenvolvimento , Bacillus anthracis/imunologia , Teorema de Bayes , Morte Celular , Simulação por Computador , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Exposição por Inalação , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Camundongos , Viabilidade Microbiana , Fagocitose , Densidade Demográfica , Esporos Bacterianos/crescimento & desenvolvimento , Esporos Bacterianos/imunologia , Processos Estocásticos , Fatores de Tempo
11.
Int Immunopharmacol ; 100: 108090, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34507108

RESUMO

BACKGROUND: Immunomodulatory therapies are claimed to enhance antimicrobial immunity and counterbalance antimicrobial resistance mechanisms of pathogenic bacteria. PURPOSE: To investigate whether caffeine can be useful for control of inflammation derived from experimental systemic infection with Listeria monocytogenes. METHODS: Peritoneal macrophages (pMØ) from Swiss mice were cultured with caffeine in 96-well plates, and then infected with virulent L. monocytogenes 619. In another experiment, the pMØ were first infected with the bacterium and then treated with caffeine. Swiss mice were inoculated intraperitoneally with L. monocytogenes and then treated intravenously with caffeine (0.05; 0.5 or 5 mg/Kg). RESULTS: Caffeine did not exert direct antibacterial activity in vitro against L. monocytogenes. Macrophages exposed to caffeine before or after infection with L. monocytogenes had increased cell viability, although the intracellular bacterial loads were similar to the control groups. Caffeine treatments of Swiss mice reduced leukocyte infiltration into the peritoneal cavity after L. monocytogenes infection. However, the bacterial burden was reduced in the spleen and liver. The mRNA expressions of IL-1ß, IL-6 and the enzyme inducible nitric oxide synthase (iNOS) were reduced whereas IL-10 was increased. CONCLUSION: Caffeine has an anti-infectious potential and ameliorated infection-derived inflammation following experimental infection with L. monocytogenes.


Assuntos
Anti-Inflamatórios/farmacologia , Cafeína/farmacologia , Inflamação/tratamento farmacológico , Listeria monocytogenes/patogenicidade , Listeriose/tratamento farmacológico , Macrófagos Peritoneais/efeitos dos fármacos , Animais , Cafeína/análogos & derivados , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Mediadores da Inflamação/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/metabolismo , Listeriose/microbiologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Virulência
12.
Inflammation ; 44(6): 2429-2447, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34505975

RESUMO

Gut microbiota contribute to shaping the immune repertoire of the host, whereas probiotics may exert beneficial effects by modulating immune responses. Having in mind the differences in both the composition of gut microbiota and the immune response between rats of Albino Oxford (AO) and Dark Agouti (DA) rat strains, we investigated if intraperitoneal (i.p.) injection of live Lactobacillus rhamnosus (LB) may influence peritoneal cavity cell response to in vitro treatments with selected microbiota in the rat strain-dependent manner. Peritoneal cavity cells from AO and DA rats were lavaged two (d2) and seven days (d7) following i.p. injection with LB and tested for NO, urea, and H2O2 release basally, or upon in vitro stimulation with autologous E.coli and Enterococcus spp. Whereas the single i.p. injection of LB nearly depleted resident macrophages and increased the proportion of small inflammatory macrophages and monocytes on d2 in both rat strains, greater proportion of MHCIIhiCD163- and CCR7+ cells and increased NO/diminished H2O2 release in DA compared with AO rats suggest a more intense inflammatory priming by LB in this rat strain. Even though E.coli- and/or Enterococcus spp.-induced rise in H2O2 release in vitro was abrogated by LB in cells from both rat strains, LB prevented microbiota-induced increase in NO/urea ratio only in cells from AO and augmented it in cells from DA rats. Thus, the immunomodulatory properties may not be constant for particular probiotic bacteria, but shaped by innate immunity of the host.


Assuntos
Microbioma Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunidade Inata , Lacticaseibacillus rhamnosus/imunologia , Macrófagos Peritoneais/microbiologia , Cavidade Peritoneal/microbiologia , Probióticos , Animais , Citocinas/metabolismo , Enterococcus/imunologia , Escherichia coli/imunologia , Feminino , Interações Hospedeiro-Patógeno , Peróxido de Hidrogênio/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Óxido Nítrico/metabolismo , Fenótipo , Ratos , Especificidade da Espécie , Ureia/metabolismo
13.
Cell Rep ; 35(3): 109012, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33882312

RESUMO

Caspase-11 sensing of intracellular lipopolysaccharide (LPS) plays critical roles during infections and sepsis. However, the key cell types that sense intracellular LPS and their contributions to the host responses at the organismal level are not completely clear. Here, we show that macrophage/monocyte-specific caspase-11 plays a dominant role in mediating the pathological manifestations of endotoxemia, including gasdermin D (GSDMD) activation, interleukin (IL)-1ß, IL-18, and damage-associated molecular pattern (DAMP) release, tissue damage, and death. Surprisingly, caspase-11 expression in CD11c+ cells and intestinal epithelial cells (IECs) plays minor detrimental roles in LPS shock. In contrast, caspase-11 expression in neutrophils is dispensable for LPS-induced lethality. Importantly, caspase-11 sensing of intracellular LPS in LyzM+ myeloid cells and MRP8+ neutrophils, but not CD11c+ cells and IECs, is necessary for bacterial clearance and host survival during intracellular bacterial infection. Thus, we reveal hierarchical cell-type-specific roles of caspase-11 that govern the host-protective and host-detrimental functions of the cytosolic LPS surveillance.


Assuntos
Caspases Iniciadoras/genética , Lipopolissacarídeos/toxicidade , Macrófagos Peritoneais/imunologia , Neutrófilos/imunologia , Choque Séptico/imunologia , Baço/imunologia , Animais , Burkholderia/crescimento & desenvolvimento , Burkholderia/patogenicidade , Antígenos CD11/genética , Antígenos CD11/imunologia , Calgranulina A/genética , Calgranulina A/imunologia , Caspases Iniciadoras/imunologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Feminino , Regulação da Expressão Gênica , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Fígado/imunologia , Fígado/microbiologia , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/imunologia , Monócitos/imunologia , Monócitos/microbiologia , Neutrófilos/microbiologia , Proteínas de Ligação a Fosfato/genética , Proteínas de Ligação a Fosfato/imunologia , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/imunologia , Choque Séptico/genética , Choque Séptico/microbiologia , Choque Séptico/mortalidade , Transdução de Sinais , Baço/microbiologia , Análise de Sobrevida
14.
Am J Respir Cell Mol Biol ; 64(5): 641-643, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33929292
15.
J Antibiot (Tokyo) ; 74(4): 244-254, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33479520

RESUMO

Therapeutic strategies that target bacterial virulence have received considerable attention. The type III secretion system (T3SS) is important for bacterial virulence and represents an attractive therapeutic target. Recently, we developed a new small-molecule inhibitor belonging to a class 2,4-disubstituted-4H-[1,3,4]-thiadiazine-5-ones, Fluorothiazinon (FT-previously called CL-55). FT effectively suppressed T3SS of Chlamydia spp., Pseudomonas aeruginosa, and Salmonella without affecting bacterial growth in vitro. FT was previously characterized by low toxicity, stability, and therapeutic efficacy in animal models. Salmonella T3SS inhibition by FT was studied using in vitro assays for effector proteins detection and estimation of salmonella replication in peritoneal macrophages. The antibacterial effect of FT in vivo was investigated in murine models of salmonella chronic systemic and acute infection. Oral administration of the virulent strain of Salmonella enterica serovar Typhimurium to mice-induced chronic systemic infection with the pathogen persistence in different lymphoid organs such as spleens, Peyer's plaques, and mesenteric lymph nodes. We found that FT suppressed orally induced salmonella infection both with therapeutic and prophylactic administration. Treatment by FT at a dose of 50 mg/kg for 4 days starting from day 7 post-infection (therapy) as well as for 4 days before infection (prevention) led to practically complete eradication of salmonella in mice. FT shows a strong potential for antibacterial therapy and could be used as a substance in the design of antibacterial drugs for pharmaceutical intervention including therapy of antibiotic-resistant infections.


Assuntos
Anilidas/farmacologia , Antibacterianos/farmacologia , Infecções por Salmonella/tratamento farmacológico , Tiadiazinas/farmacologia , Ampicilina/farmacologia , Anilidas/administração & dosagem , Anilidas/farmacocinética , Animais , Antibacterianos/administração & dosagem , Antibacterianos/farmacocinética , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos Endogâmicos , Coelhos , Intoxicação Alimentar por Salmonella/tratamento farmacológico , Intoxicação Alimentar por Salmonella/prevenção & controle , Infecções por Salmonella/prevenção & controle , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Tiadiazinas/administração & dosagem , Tiadiazinas/farmacocinética , Distribuição Tecidual , Sistemas de Secreção Tipo III/antagonistas & inibidores
16.
Braz J Microbiol ; 52(1): 19-31, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32772310

RESUMO

Melanin is a Sporothrix virulence factor that can inhibit the innate immune functions of macrophages such as phagocytosis and killing. However, no data on melanin's influence on antigen presentation by macrophages are available. In this study, we used conidia, yeasts, and melanin ghosts (MGs) from a black Sporothrix globosa strain (MEL+) and its ultraviolet-induced albino mutant (MEL-), to study the influence of melanin on expression of molecules involved in antigen presentation by mouse macrophages (MHC class II, CD80, CD86), as well as on levels of transcription factors regulating their expression (CIITA and promoters I, III, and IV). A murine infection model was used to assess the virulence of both strains and differences in expression of MHC class II and CD80/86 in vivo. MHC class II, CD86 CIITA, and PIV expressions were lower in macrophages infected with MEL+ than in macrophages infected with MEL- conidia, while CD80 expression was similar. No statistical difference in gene expression was observed between macrophages infected by MEL+ and MEL- yeasts. Infection by MGs alone had no clear effect on expression of antigen presentation-associated molecules. Mice infected with MEL+ S. globosa had significantly higher fungal burdens in the lung, liver, spleen, kidney, and testicle compared with mice infected with MEL- S. globosa 21 days post-infection. MHC class II expression changes in the animal study were similar to those observed in the in vitro experiment. Our results indicate that S. globosa melanin can inhibit expression of antigen presentation-associated molecules during both the early and late stages of infection, representing a new mechanism to evade host immunity and to enhance dissemination. Further investigations of melanin's impact on adaptive immunity will be helpful in understanding this fungal virulence factor.


Assuntos
Macrófagos Peritoneais/imunologia , Melaninas/imunologia , Sporothrix/imunologia , Esporotricose/microbiologia , Animais , Apresentação de Antígeno , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Interações Hospedeiro-Patógeno , Humanos , Fígado/microbiologia , Pulmão/microbiologia , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Sporothrix/genética , Esporotricose/genética , Esporotricose/imunologia
17.
Immunol Invest ; 50(6): 700-725, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32602757

RESUMO

Anti-cytokine therapy is widely acknowledged as an anti-inflammatory technique to treat varied infectious diseases. TNF-α and IL-1ß are major cytokines that regulate every aspect of the inflammatory process. However, the effects of single or dual cytokine neutralization on S. aureus mediated CXCL8 secretion and CXCR1 expression in murine peritoneal macrophages remained noninvestigated. Thus we aimed to explore the effects of kinetic-dose dependent neutralization of TNF-α and IL-1ß using specific anti-cytokine antibodies and its influential impact on the CXCL8/CXCR1 axis at different stages of S. aureus (30, 60, and 90 min) infection. The murine peritoneal macrophages were isolated and infected with viable S. aureus followed by subsequent addition of anti-TNF-α and anti-IL-1ß into the medium. The treated cells were centrifuged and lysate and supernatant collected for various experiments. The ROS generation was measured and cytokine production was estimated by ELISA. The expression of TNFR1, IL-1R, CXCR1, signaling molecules (NF-κB and JNK) were evaluated by Western blot. The role of single or dual cytokine neutralization on intracellular bacterial phagocytosis had also been analyzed by confocal microscopy. Dual cytokine neutralization significantly suppressed ROS, cytokines, CXCL8 secretion, and intracellular bacterial count compared to single cytokine neutralization and it was more apparent at 90 min post S. aureus infection. There was a drastic reduction in TNFR1, IL-1R, and CXCR1 expression on macrophage surface due to reduced expression of downstream signaling molecules, NF-κB and JNK. Hence dual cytokine neutralization was more effectual compared to single cytokine neutralization in the downregulation of S. aureus induced CXCR1 expression.


Assuntos
Imunossupressores/farmacologia , Interleucina-1beta/antagonistas & inibidores , Interleucina-8/metabolismo , Infecções Estafilocócicas/tratamento farmacológico , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Carga Bacteriana/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Humanos , Imunossupressores/uso terapêutico , Interleucina-1beta/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Cultura Primária de Células , Receptores de Interleucina-8A/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Fator de Necrose Tumoral alfa/metabolismo
18.
Front Immunol ; 11: 573550, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33362762

RESUMO

GPI-anchored uPAR is the receptor for the extracellular serine protease urokinase-type plasminogen activator (uPA). Though uPAR role in inflammatory processes is documented, underlying mechanisms are not fully understood. In this study we demonstrate that uPAR is a part of Toll-like receptor 4 (TLR4) interactome. Downregulation of uPAR expression resulted in diminished LPS-induced TLR4 signaling, less activation of NFκB, and decreased secretion of inflammatory mediators in myeloid and non-myeloid cells in vitro. In vivo uPAR-/- mice demonstrated better survival, strongly diminished inflammatory response and better organ functions in cecal ligation and puncture mouse polymicrobial sepsis model. Mechanistically, GPI-uPAR and soluble uPAR colocalized with TLR4 on the cell membrane and interacted with scavenger receptor CD36. Our data show that uPAR can interfere with innate immunity response via TLR4 and this mechanism represents a potentially important target in inflammation and sepsis therapy.


Assuntos
Células Epiteliais/efeitos dos fármacos , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sepse/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Antígenos CD36/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Humanos , Inflamação/genética , Inflamação/microbiologia , Inflamação/prevenção & controle , Mediadores da Inflamação/metabolismo , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Células RAW 264.7 , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Sepse/genética , Sepse/microbiologia , Sepse/prevenção & controle , Transdução de Sinais , Receptor 4 Toll-Like/genética
19.
Bull Exp Biol Med ; 169(4): 467-469, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32910376

RESUMO

Cultured peritoneal macrophages from intact (control) and BCG-infected (experiment) male BALB/c mice were studied 90 days after infection. Polarization of macrophages by M1 (expression of GM-CSF, IFNγ, and CD16/32) and M2 (expression of bFGF and CD36) differentiation pathways was studied with consideration for their the nuclearity class. Mononuclear cells predominated (90% and higher) in macrophage cultures of both groups and presumably, were presented by mainly epithelioid cells. The results indicated polarization of mononuclear and multinuclear macrophages in the M2 direction under conditions of BCG granulomatosis and a higher initial M2 polarization of binuclear macrophages. In control cultures, the ratio of M2 to M1 macrophages was 0.57, in experimental cultures this ratio was 1.6. It seems that long persistence of Mycobacterium tuberculosis in macrophages served as a factor stimulating the plastic processes and transformation of macrophages into epithelioid cells that form the "core" of granulomas and their enlargement upon incorporation of macrophages.


Assuntos
Células Epitelioides/patologia , Regulação da Expressão Gênica/imunologia , Macrófagos Peritoneais/patologia , Mycobacterium bovis/crescimento & desenvolvimento , Tuberculose/patologia , Animais , Antígenos CD36/genética , Antígenos CD36/imunologia , Diferenciação Celular , Transdiferenciação Celular/genética , Transdiferenciação Celular/imunologia , Células Epitelioides/imunologia , Células Epitelioides/microbiologia , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Interferon gama/genética , Interferon gama/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium bovis/patogenicidade , Cultura Primária de Células , Receptores de IgG/genética , Receptores de IgG/imunologia , Tuberculose/genética , Tuberculose/imunologia , Tuberculose/microbiologia
20.
J Clin Invest ; 130(11): 5858-5874, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32759503

RESUMO

Mitochondria have emerged as key actors of innate and adaptive immunity. Mitophagy has a pivotal role in cell homeostasis, but its contribution to macrophage functions and host defense remains to be delineated. Here, we showed that lipopolysaccharide (LPS) in combination with IFN-γ inhibited PINK1-dependent mitophagy in macrophages through a STAT1-dependent activation of the inflammatory caspases 1 and 11. In addition, we demonstrated that the inhibition of mitophagy triggered classical macrophage activation in a mitochondrial ROS-dependent manner. In a murine model of polymicrobial infection (cecal ligature and puncture), adoptive transfer of Pink1-deficient bone marrow or pharmacological inhibition of mitophagy promoted macrophage activation, which favored bactericidal clearance and led to a better survival rate. Reciprocally, mitochondrial uncouplers that promote mitophagy reversed LPS/IFN-γ-mediated activation of macrophages and led to immunoparalysis with impaired bacterial clearance and lowered survival. In critically ill patients, we showed that mitophagy was inhibited in blood monocytes of patients with sepsis as compared with nonseptic patients. Overall, this work demonstrates that the inhibition of mitophagy is a physiological mechanism that contributes to the activation of myeloid cells and improves the outcome of sepsis.


Assuntos
Bactérias/imunologia , Ativação de Macrófagos , Macrófagos Peritoneais/imunologia , Mitofagia/imunologia , Sepse/imunologia , Animais , Feminino , Humanos , Interferon gama/imunologia , Lipopolissacarídeos/imunologia , Macrófagos Peritoneais/microbiologia , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Proteínas Quinases/imunologia , Células RAW 264.7 , Sepse/microbiologia , Sepse/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA