Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.772
Filtrar
1.
J Ethnopharmacol ; 336: 118661, 2025 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-39159837

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Shuangdan Jiedu Decoction (SJD) is a formula composed of six Chinese herbs with heat-removing and detoxifying, antibacterial, and anti-inflammatory effects, which is clinically used in the therapy of various inflammatory diseases of the lungs including COVID-19, but the therapeutic material basis of its action as well as its molecular mechanism are still unclear. AIM OF THE STUDY: The study attempted to determine the therapeutic effect of SJD on LPS-induced acute lung injury (ALI), as well as to investigate its mechanism of action and assess its therapeutic potential for the cure of inflammation-related diseases in the clinical setting. MATERIALS AND METHODS: We established an ALI model by tracheal drip LPS, and after the administration of SJD, we collected the bronchoalveolar lavage fluid (BALF) and lung tissues of mice and examined the expression of inflammatory factors in them. In addition, we evaluated the effects of SJD on the cyclic guanosine monophosphate-adenosine monophosphate synthase -stimulator of interferon genes (cGAS-STING) and inflammasome by immunoblotting and real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS: We demonstrated that SJD was effective in alleviating LPS-induced ALI by suppressing the levels of pro-inflammatory cytokines in the BALF, improving the level of lung histopathology and the number of neutrophils, as well as decreasing the inflammatory factor-associated gene expression. Importantly, we found that SJD could inhibit multiple stimulus-driven activation of cGAS-STING and inflammasome. Further studies showed that the Chinese herbal medicines in SJD had no influence on the cGAS-STING pathway and inflammasome alone at the formulated dose. By increasing the concentration of these herbs, we observed inhibitory effects on the cGAS-STING pathway and inflammasome, and the effect exerted was maximal when the six herbs were combined, indicating that the synergistic effects among these herbs plays a crucial role in the anti-inflammatory effects of SJD. CONCLUSIONS: Our research demonstrated that SJD has a favorable protective effect against ALI, and its mechanism of effect may be associated with the synergistic effect exerted between six Chinese medicines to inhibit the cGAS-STING and inflammasome abnormal activation. These results are favorable for the wide application of SJD in the clinic as well as for the development of drugs for ALI from herbal formulas.


Assuntos
Lesão Pulmonar Aguda , Medicamentos de Ervas Chinesas , Inflamassomos , Lipopolissacarídeos , Proteínas de Membrana , Nucleotidiltransferases , Transdução de Sinais , Animais , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Lipopolissacarídeos/toxicidade , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Nucleotidiltransferases/metabolismo , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos , Masculino , Transdução de Sinais/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Modelos Animais de Doenças , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/metabolismo , Líquido da Lavagem Broncoalveolar/citologia
2.
Sci Rep ; 14(1): 26216, 2024 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-39482340

RESUMO

NLRP3 inflammasomes- pyroptosis axis is activated by microcirculation dysfunction and touched off severe acute pancreatitis (SAP). Activation of PGC-1α can improve microcirculation dysfunction by promoting mitochondrial biogenesis. Resveratrol (RSV), one typical SIRT1 agonist, possesses the ability of alleviating SAP and activing PGC-1α. Therefore, the study was designated to explore whether the protective effect of RSV in SAP was though suppressing NLRP3 inflammasomes- pyroptosis axis via advancing SIRT1/PGC-1α-dependent mitochondrial biogenesis. The models of SAP were induced by treating with sodium taurodeoxycholate in rats and AR42J cells. The pathological injury, water content (dry/wet ratio) and microcirculation function of pancreas, activity of lipase and amylase were used to evaluate pancreatic damage. The expression of inflammatory cytokine was measured by ELISA and RT-PCR. The damage of mitochondrial was evaluated by measuring the changes in Mitochondrial Membrane Potential (ΔΨm), mitochondrial ROS, ATP content and MDA as well as relocation of mtDNA and the activity of SOD and GSH. The expressions of NLRP3 inflammasomes- pyroptosis axis proteins were detected by Western blotting as well as SIRT1/PGC-1α/NRF1/TFAM pathway protein. Moreover, the modification of PGC-1α was measured by co-immunoprecipitation. The results displayed that RSV can significantly improve the damage of pancreas and mitochondrial, decrease the expression of pro-inflammatory factor and the activation of NLRP3 inflammasomes- pyroptosis axis, promote the expression of an-inflammatory factor and the deacetylation of PGC-1α together with facilitating SIRT1/PGC-1α-mediating mitochondrial biogenesis. Therefore, the protective effect of RSV in SAP is though inactivation of NLRP3 inflammasomes- pyroptosis axis via promoting mitochondrial biogenesis in a SIRT1/PGC-1α-dependent manner.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Biogênese de Organelas , Pancreatite , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Resveratrol , Transdução de Sinais , Sirtuína 1 , Sirtuína 1/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Animais , Resveratrol/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Masculino , Pancreatite/tratamento farmacológico , Pancreatite/metabolismo , Pancreatite/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Ratos Sprague-Dawley , Piroptose/efeitos dos fármacos , Linhagem Celular
3.
Nat Commun ; 15(1): 9035, 2024 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-39426974

RESUMO

Localized delivery of inflammasome inhibitors in phagocytic macrophages could be promising for psoriasis treatment. The present work demonstrates the development of non-spherical lipid nanoparticles, mimicking pathogen-like shapes, consisting of an anti-inflammatory inflammasome inhibiting lipid (pyridoxine dipalmitate) as a trojan horse. The nanorods inhibit inflammasome by 3.8- and 4.5-fold compared with nanoellipses and nanospheres, respectively. Nanorods reduce apoptosis-associated speck-like protein and lysosomal rupture, restrain calcium influx, and mitochondrial reactive oxygen species. Dual inflammasome inhibitor (NLRP3/AIM-2-IN-3) loaded nanorods cause synergistic inhibition by 21.5- and 59-folds compared with nanorods and free drug, respectively alongside caspase-1 inhibition. The NLRP3/AIM-2-IN-3 nanorod when transformed into a polymeric scaffold, simultaneously and effectively inhibits RNA levels of NLRP3, AIM2, caspase-1, chemokine ligand-2, gasdermin-D, interleukin-1ß, toll-like receptor 7/ 8, and IL-17A by 6.4-, 1.6-, 2.0-, 13.0-, 4.2-, 24.4-, 4.3-, and 1.82-fold, respectively in psoriatic skin in comparison to Imiquimod positive control group in an in-vivo psoriasis-like mice model.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nanotubos , Psoríase , Psoríase/tratamento farmacológico , Animais , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Camundongos , Humanos , Nanotubos/química , Polímeros/química , Nanopartículas/química , Espécies Reativas de Oxigênio/metabolismo , Modelos Animais de Doenças , Lipídeos/química , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Caspase 1/metabolismo , Feminino , Masculino , Lipossomos
4.
Mediators Inflamm ; 2024: 3938136, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39381066

RESUMO

Purpose: Aloperine (ALO), an alkaloid isolated from Sophora alopecuroides L., possesses multiple pharmacological activities and holds a promise potential for the treatment of various clinical conditions, including skin hypersensitivity, cancer, and inflammatory disorders. The purpose of this study was to investigate the role of ALO in acetaminophen (N-acetyl-para-aminophenol (APAP))-induced acute liver injury and its underlying mechanisms. Materials and Methods: An animal model of acute liver injury was induced by intraperitoneal injection of APAP (150 mg/kg). Prior to APAP injection, ALO (40 mg/kg) was administered daily for 7 consecutive days. Serum alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase levels were then measured using an automated chemical analyzer. Histopathological changes were evaluated using hematoxylin and eosin staining. Oxidative stress levels were measured by detecting superoxide dismutase (SOD), glutathione (GSH), and malondialdehyde (MDA). Pro-inflammatory cytokines were detected in serum and liver tissues using ELISA and quantitative real-time polymerase chain reaction (q-PCR). The expression of members of the HMGB1/TLR4/NF-κB signaling pathway and NLRP3 inflammasome were determined by Western blot and/or q-PCR. In addition, the expression and location of NLRP3, cleaved caspase-1, high-mobility group box 1 (HMGB1), and phosphorylated p65 (p-p65) were detected by immunofluorescence. Results: Pretreatment with ALO significantly protected mice from APAP-induced acute liver injury, with decreased MDA content, and significantly increased GSH and SOD activities. Furthermore, ALO pretreatment reduced the release of pro-inflammatory cytokines (IL-1ß and TNF-α) and decreased the expression of caspase-1, cleaved caspase-1, and NLRP3. In addition, ALO pretreatment also inhibited the activation of the HMGB1/TLR4/NF-κB signaling pathway. Conclusion: Taken together, ALO can ameliorate APAP-induced acute liver injury by inhibiting oxidative stress, inflammation by inhibiting the HMGB1/TLR4/NF-κB, and NLRP3/inflammasome pathway.


Assuntos
Acetaminofen , Doença Hepática Induzida por Substâncias e Drogas , Proteína HMGB1 , Inflamassomos , NF-kappa B , Proteína 3 que Contém Domínio de Pirina da Família NLR , Piperidinas , Transdução de Sinais , Receptor 4 Toll-Like , Animais , Receptor 4 Toll-Like/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Camundongos , Acetaminofen/efeitos adversos , NF-kappa B/metabolismo , Masculino , Proteína HMGB1/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Inflamassomos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Fígado/efeitos dos fármacos , Fígado/metabolismo , Quinolizidinas
5.
Front Cell Infect Microbiol ; 14: 1384072, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39376663

RESUMO

There is evidence that in infected cells in vitro the meningococcal HrpA/HrpB two-partner secretion system (TPS) mediates the exit of bacteria from the internalization vacuole and the docking of bacteria to the dynein motor resulting in the induction of pyroptosis. In this study we set out to study the role of the HrpA/HrpB TPS in establishing meningitis and activating pyroptotic pathways in an animal model of meningitis using a reference serogroup C meningococcal strain, 93/4286, and an isogenic hrpB knockout mutant, 93/4286ΩhrpB. Survival experiments confirmed the role of HrpA/HrpB TPS in the invasive meningococcal disease. In fact, the ability of the hrpB mutant to replicate in brain and spread systemically was impaired in mice infected with hrpB mutant. Furthermore, western blot analysis of brain samples during the infection demonstrated that: i. N. meningitidis activated canonical and non-canonical inflammasome pyroptosis pathways in the mouse brain; ii. the activation of caspase-11, caspase-1, and gasdermin-D was markedly reduced in the hrpB mutant; iii. the increase in the amount of IL-1ß and IL-18, which are an important end point of pyroptosis, occurs in the brains of mice infected with the wild-type strain 93/4286 and is strongly reduced in those infected with 93/4286ΩhrpB. In particular, the activation of caspase 11, which is triggered by cytosolic lipopolysaccharide, indicates that during meningococcal infection pyroptosis is induced by intracellular infection after the exit of the bacteria from the internalizing vacuole, a process that is hindered in the hrpB mutant. Overall, these results confirm, in an animal model, that the HrpA/HrpB TPS plays a role in the induction of pyroptosis and suggest a pivotal involvement of pyroptosis in invasive meningococcal disease, paving the way for the use of pyroptosis inhibitors in the adjuvant therapy of the disease.


Assuntos
Encéfalo , Caspase 1 , Modelos Animais de Doenças , Meningite Meningocócica , Neisseria meningitidis , Piroptose , Animais , Neisseria meningitidis/patogenicidade , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Camundongos , Meningite Meningocócica/microbiologia , Caspase 1/metabolismo , Encéfalo/patologia , Encéfalo/microbiologia , Encéfalo/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Caspases/metabolismo , Caspases Iniciadoras/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Proteínas de Ligação a Fosfato/genética , Interleucina-1beta/metabolismo , Inflamassomos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Sistemas de Secreção Bacterianos/genética , Feminino , Interleucina-18/metabolismo , Gasderminas
6.
PLoS One ; 19(10): e0311624, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39388398

RESUMO

Tilianin (TIL), a bioactive component derived from Dracocephalum Moldavica L., has been recognized for its anti-inflammatory properties. However, its effects on the Nlrp3 inflammasome within endothelial progenitor cells (EPCs) during myocardial ischemia-reperfusion injury (MIRI) remain unexplored. This study aimed to elucidate the role of TIL in modulating Nlrp3 inflammasome activation under MIRI conditions. A mouse model of MIRI was established to assess the therapeutic potential of TIL. EPCs treated with TIL at concentrations of 5, 10, and 20 µM were administered into the myocardium before reperfusion. Additionally, the cardioprotective effects of TIL were further examined by pre-treating EPCs with the compound before exposing them to hypoxia/reoxygenation (H/R) using cardiomyocyte supernatants. The impact on Nlrp3 inflammasome was assessed through western blotting, immunofluorescence, and ELISA. Our results showed that TIL concentration-dependently inhibited Nlrp3 inflammasome-related protein levels,and inhibited Asc oligomerization and Asc-Speck complex formation in EPCs, resulting in improved the migratory capacity and vascular structure formation of EPCs. In addition, TIL-treated EPCs significantly attenuated I/R injury and improved cardiac function. These results suggest that TIL ameliorates the inflammatory response in EPCs by suppressing Nlrp3 inflammasome activation, thereby facilitating neovascularization in the myocardium and conferring protection against MIRI. The study provides valuable insights into the potential of TIL as a therapeutic agent for cardiovascular diseases linked to ischemia-reperfusion injury.


Assuntos
Células Progenitoras Endoteliais , Glicosídeos , Inflamassomos , Traumatismo por Reperfusão Miocárdica , Proteína 3 que Contém Domínio de Pirina da Família NLR , Animais , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Células Progenitoras Endoteliais/efeitos dos fármacos , Células Progenitoras Endoteliais/metabolismo , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Masculino , Glicosídeos/farmacologia , Camundongos Endogâmicos C57BL , Flavonoides/farmacologia , Modelos Animais de Doenças , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo
7.
Sci Rep ; 14(1): 23882, 2024 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-39396074

RESUMO

Beyond its clinical diversity and severity, acute myeloid leukemia (AML) is known for its complex molecular background and for rewiring biological processes to aid disease onset and maintenance. FLT3 mutations are among the most recurring molecular entities that cooperatively drive AML, and their inhibition is a critical molecularly oriented therapeutic strategy. Despite being a promising avenue, it still faces challenges such as intrinsic and acquired drug resistance, which led us to investigate whether and how autophagy and inflammasome interact and whether this interaction could be leveraged to enhance FLT3 inhibition as a therapeutic strategy. We observed a strong and positive correlation between the expression of key genes associated with autophagy and the inflammasome. Gene set enrichment analysis of the FLT3-ITD samples and their ex vivo response to five different FLT3 inhibitors revealed a common molecular signature compatible with autophagy and inflammasome activation across all poor responders. Inflammasome activation was also shown to strongly increase the likelihood of a poor ex vivo response to the FLT3 inhibitors quizartinib and sorafenib. These findings reveal a distinct molecular pattern within FLT3-ITD AML samples that underscores the necessity for further exploration into how approaching these supportive parallel yet altered pathways could improve therapeutic strategies.


Assuntos
Autofagia , Inflamassomos , Leucemia Mieloide Aguda , Inibidores de Proteínas Quinases , Tirosina Quinase 3 Semelhante a fms , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Autofagia/efeitos dos fármacos , Inflamassomos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Mutação , Feminino , Masculino , Benzotiazóis/farmacologia , Pessoa de Meia-Idade , Sorafenibe/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Idoso , Compostos de Fenilureia
8.
Front Immunol ; 15: 1419117, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39399507

RESUMO

Background: Dysregulated innate immune responses underlie multiple inflammatory diseases, but clinical translation of preclinical innate immunity research in mice is hampered by the difficulty of studying human inflammatory reactions in an in vivo context. We therefore sought to establish in vivo human inflammatory responses in NSG-QUAD mice that express four human myelopoiesis transgenes to improve engraftment of a human innate immune system. Methods: We reconstituted NSG-QUAD mice with human hematopoietic stem and progenitor cells (HSPCs), after which we evaluated human myeloid cell development and subsequent human responses to systemic and local lipopolysaccharide (LPS) challenges. Results: NSG-QUAD mice already displayed engraftment of human monocytes, dendritic cells and granulocytes in peripheral blood, spleen and liver at 6 weeks after HSPC reconstitution, in which both classical, intermediate and non-classical monocytes were present. These huNSG-QUAD mice responded to intraperitoneal and intranasal LPS challenges with production of NF-κB-dependent human cytokines, a human type I interferon response, as well as inflammasome-mediated production of human IL-1ß and IL-18. The latter were specifically abrogated by the NLRP3 inhibitor MCC950, while LPS-induced human monocyte death was not altered. Besides providing proof-of-principle for small molecule testing of human inflammatory reactions in huNSG-QUAD mice, this observation suggests that LPS-induced in vivo release of human NLRP3 inflammasome-generated cytokines occurs in a cell death-independent manner. Conclusion: HuNSG-QUAD mice are competent for the NF-κB, interferon and inflammasome effectors of human innate immunity, and can thus be utilized to investigate signaling mechanisms and pharmacological targeting of human inflammatory responses in an in vivo setting.


Assuntos
Imunidade Inata , Lipopolissacarídeos , Mielopoese , Animais , Humanos , Mielopoese/imunologia , Mielopoese/genética , Camundongos , Transgenes , Camundongos Transgênicos , Inflamassomos/metabolismo , Inflamassomos/imunologia , Citocinas/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Modelos Animais de Doenças , Inflamação/imunologia , Inflamação/genética
9.
J Neuroinflammation ; 21(1): 261, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39407211

RESUMO

The NLR family pyrin domain containing 3 (NLRP3) inflammasome in microglia is intimately linked to the pathogenesis of Alzheimer's disease (AD). Although NLRP3 inflammasome activity is regulated by cellular metabolism, the underlying mechanism remains elusive. Here, we found that under the pathological conditions of AD, the activation of NLRP3 inflammasome in microglia is accompanied by increased glutamine metabolism. Suppression of glutaminase, the rate limiting enzyme in glutamine metabolism, attenuated the NLRP3 inflammasome activation both in the microglia of AD mice and cultured inflammatory microglia. Mechanistically, inhibiting glutaminase blocked the anaplerotic flux of glutamine to the tricarboxylic acid cycle and amino acid synthesis, down-regulated mTORC1 signaling by phosphorylating AMPK, which stimulated mitophagy and limited the accumulation of intracellular reactive oxygen species, ultimately prevented the activation of NLRP3 inflammasomes in activated microglia during AD. Taken together, our findings suggest that glutamine metabolism regulates the activation of NLRP3 inflammasome through mitophagy in microglia, thus providing a potential therapeutic target for AD treatment.


Assuntos
Doença de Alzheimer , Glutamina , Inflamassomos , Microglia , Mitofagia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Animais , Humanos , Masculino , Camundongos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Células Cultivadas , Glutamina/metabolismo , Inflamassomos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Mitofagia/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Feminino
10.
CNS Neurosci Ther ; 30(10): e70061, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39392762

RESUMO

BACKGROUND: Inflammatory and immune responses in the brain that contribute to various neuropsychiatric disorders may begin as microglial "priming". Interferon (IFN)-γ is known to cause microglial priming, but the mechanism is unclear. METHODS: We examined the effects of IFN-γ on gene expression, microglial activation, inflammatory and immune responses and activity of the NLRP3 inflammasome in primary microglia and in the brains of mice. RESULTS: Our results showed that treating microglial cultures with IFN-γ induced a hedgehog-like morphology and upregulated markers of microglial activation (CD86, CD11b) and pro-inflammatory molecules (IL-1ß, IL-6, TNF-α, iNOS), while downregulating markers of microglial homeostasis (CX3CR1, CD200R1), anti-inflammatory molecules (MCR1, Arg-1) and neurotrophic factors (IGF-1, BDNF). IFN-γ also upregulated markers of NLRP3 inflammasome activation (NLRP3, caspase-1, gasdermin D, IL-18). This particular transcriptional profiling makes IFN-γ-primed microglia with exaggerated responses upon lipopolysaccharide (LPS) stimulation. The level of NLRP3, caspase-1, gasdermin D, IL-1ß, IL-18, TNF-α and iNOS in microglia cultures treated with both IFN-γ and LPS were highest than with either one alone. Injecting IFN-γ into the lateral ventricle of mice induced similar morphological and functional changes in hippocampal microglia as in primary microglial cultures. The effects of IFN-γ on NLRP3 inflammasome and microglia from cultures or hippocampus were abolished when STAT1 was inhibited using fludarabin. Injecting mice with IFN-γ alone or together with LPS induced anxiety- and depression-like behaviors and impaired hippocampus-dependent spatial memory; these effects were mitigated by fludarabin. CONCLUSIONS: IFN-γ primes microglia by activating STAT1, which upregulates genes that activate the NLRP3 inflammasome. Inhibiting the IFN-γ/STAT1 axis may be a way to treat neurodegenerative diseases and psychiatric disorders that involve microglial priming.


Assuntos
Inflamassomos , Interferon gama , Camundongos Endogâmicos C57BL , Microglia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fator de Transcrição STAT1 , Animais , Microglia/efeitos dos fármacos , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Interferon gama/farmacologia , Fator de Transcrição STAT1/metabolismo , Camundongos , Inflamassomos/metabolismo , Células Cultivadas , Masculino , Lipopolissacarídeos/farmacologia
11.
BMC Immunol ; 25(1): 69, 2024 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-39415131

RESUMO

BACKGROUND: Porphyromonase gingivalis (P. gingivalis) is a type of bacteria that causes periodontitis, which is strongly correlated with systemic diseases such as diabetes. However, the effect of hyperglycemia on periodontitis are unclear. The present study examined the effects of high glucose levels on the response to P. gingivalis infection. RESULTS: The expression of P. gingivalis-induced interleukin-1ß (IL-1ß) and inflammasomes increased as the glucose concentration increased. High glucose conditions suppressed P. gingivalis-induced autophagy in human acute monocytic leukemia cell line (THP-1) macrophages. Zingerone increased autophagy and alleviated P. gingivalis-induced inflammatory response in THP-1 macrophages under high glucose conditions. In addition, P. gingivalis- induced inflammation in bone marrow-derived macrophages of diabetic mice was higher than in wild-type mice, but a zingerone treatment decreased the levels. Alveolar bone loss due to a P. gingivalis infection was significantly higher in diabetic mice than in wild-type mice. CONCLUSIONS: High-glucose conditions aggravated the inflammatory response to P. gingivalis infection by suppressing of autophagy, suggesting that autophagy induction could potentially to treat periodontitis in diabetes. Zingerone has potential use as a treatment for periodontal inflammation induced by P. gingivalis in diabetes patients.


Assuntos
Autofagia , Infecções por Bacteroidaceae , Glucose , Macrófagos , Periodontite , Porphyromonas gingivalis , Autofagia/efeitos dos fármacos , Animais , Humanos , Camundongos , Periodontite/imunologia , Periodontite/microbiologia , Macrófagos/imunologia , Glucose/metabolismo , Células THP-1 , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/complicações , Interleucina-1beta/metabolismo , Inflamação/imunologia , Diabetes Mellitus Experimental/imunologia , Guaiacol/análogos & derivados , Guaiacol/farmacologia , Camundongos Endogâmicos C57BL , Inflamassomos/metabolismo , Inflamassomos/imunologia , Masculino
12.
Nutrients ; 16(19)2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39408295

RESUMO

BACKGROUND/OBJECTIVES: Isatidis Folium (IF) has been used in traditional medicine for various ailments, and recent research highlights its anti-inflammatory, antiviral, and detoxifying properties. This study investigated the anti-inflammatory effects of a hydroethanolic extract of IF (EIF) on inflammasomes and colitis. METHODS: Dextran sulfate sodium (DSS)-induced colitis model C57BL/6 mice were treated with DSS, mesalamine, or EIF (200 mg/kg). Parameters such as daily disease activity index (DAI), spleen weight, colon length, and histopathology were evaluated. Intestinal fibrosis, mucin, and tight junction proteins were assessed using Masson's trichrome, periodic acid-Schiff, and immunohistochemistry staining. RAW264.7 and J774a.1 macrophages were treated with EIF and lipopolysaccharide, with cell viability assessed via the cell counting kit-8 assay, nitric oxide (NO) production with Griess reagent, and cytokine levels with the enzyme-linked immunosorbent assay. NF-κB inhibition was analyzed using the luciferase assay, and phytochemical analysis was performed using UPLC-MS/MS. RESULTS: EIF mitigated weight loss, reduced DAI scores, prevented colon shortening, and attenuated mucosal damage, fibrosis, and goblet cell loss while enhancing the tight junction protein occludin. The anti-inflammatory effects of EIF in RAW264.7 cells included reduced NO production, pro-inflammatory cytokines, and NF-κB activity, along with inhibition of NLRP3 inflammasome responses in J774a.1 cells. The key constituents identified were tryptanthrin, indigo, and indirubin. CONCLUSIONS: Animal studies demonstrated the efficacy of EIF in alleviating colitis, suggesting its potential for treating inflammatory diseases.


Assuntos
Anti-Inflamatórios , Colite , Sulfato de Dextrana , Inflamassomos , Camundongos Endogâmicos C57BL , Extratos Vegetais , Animais , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Camundongos , Células RAW 264.7 , Anti-Inflamatórios/farmacologia , Extratos Vegetais/farmacologia , Masculino , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Colo/efeitos dos fármacos , Colo/patologia , Colo/metabolismo , NF-kappa B/metabolismo , Citocinas/metabolismo
13.
Microb Pathog ; 196: 106994, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39366588

RESUMO

Recent studies indicate that bacterial outer membrane vesicles (OMVs) play a significant role in bacterial virulence and pathogenicity. Streptococcus mutans (S. mutans), a principal pathogen in dental caries, secretes a substantial number of OMVs. However, the impact of S. mutans OMVs on oral health and their underlying pathogenic mechanisms remain poorly understood. Macrophages were the initial innate immune cells to respond to bacterial invaders and their products. Therefore, we purified S. mutans OMVs, which stimulated macrophages. Compared to controls, RT-PCR and ELISA analyses revealed that S. mutans OMVs significantly increased the production of IL-1ß, IL-6, TNF-α and IL-8, with IL-1ß being notably elevated. IL-1ß production and secretion are tightly regulated by the inflammasome. Western blot analyses demonstrated that S. mutans OMVs upregulated the expression of inflammasome components, including NLRP3, NLRC4, ASC and AIM2, with a marked increase in NLRP3 expression. Silencing different inflammasome components with siRNA revealed a reduction in IL-1ß secretion induced by S. mutans OMVs, particularly through NLRP3. Additionally, ATP production and K+ efflux were found to be crucial for NLRP3 activation. Prolonged stimulation with S. mutans OMVs resulted in increased lactate production and elevated expression of glycolysis-related genes Glut-1, PFKFB3, and HK I, indicating that S. mutans OMVs significantly induce macrophage glycolysis. Furthermore, S. mutans OMVs were shown to enhance biofilm formation, increase S. mutans colonisation on epithelial cells, and inhibit macrophage phagocytosis, thereby improving the survival of S. mutans in the oral cavity. In summary, S. mutans OMVs promote the survival of S. mutans in the mouth through multiple mechanisms, potentially influencing the development of dental caries.


Assuntos
Glicólise , Inflamassomos , Interleucina-1beta , Macrófagos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Streptococcus mutans , Streptococcus mutans/genética , Streptococcus mutans/metabolismo , Streptococcus mutans/imunologia , Inflamassomos/metabolismo , Macrófagos/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Interleucina-1beta/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Camundongos , Animais , Citocinas/metabolismo , Membrana Externa Bacteriana/metabolismo , Humanos , Cárie Dentária/microbiologia
14.
Gut Microbes ; 16(1): 2413372, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39428744

RESUMO

The host restricts Salmonella enterica serovar Typhimurium infection of the gut via inflammasome-dependent sloughing of infected epithelial cells. Here we determined that concurrent caspase 1/11-dependent release of the goblet cell-derived mucin, Muc2, into the intestinal lumen also controls Salmonella burdens in infected mice. The increased release of mucins from goblet cells in the cecum and nearby proximal colon, and the subsequent thickening of the protective mucus barrier layer in the distal colon, were all dependent on the cytokines interleukin (IL)-18 and IL-22, as deficiencies in either cytokine resulted in reduced mucin secretion. Supplementation of IL-18 into IL-22 deficient mice restored mucin secretion, indicating that IL-22 acted upstream of IL-18 secretion during infection. In contrast, IL-18 and IL-22 independent signaling through Nlrp6 underlies only a modest, infection-induced increase in mucin secretion from goblet cells in the distal colon. These findings reveal that inflammasome signaling orchestrates multiple levels of protection centered on the intestinal epithelium, including pyroptosis and expulsion of infected enterocytes, as well as the release of mucins by goblet cells in the cecum and along the length of the colon. Our studies underscore the pivotal, multi-faceted role of inflammasome signaling in promoting host defense at the intestinal mucosal surface.


Assuntos
Células Caliciformes , Inflamassomos , Interleucina-18 , Interleucina 22 , Interleucinas , Mucosa Intestinal , Camundongos Endogâmicos C57BL , Mucina-2 , Salmonella typhimurium , Animais , Inflamassomos/metabolismo , Inflamassomos/imunologia , Salmonella typhimurium/imunologia , Camundongos , Células Caliciformes/metabolismo , Mucina-2/metabolismo , Mucina-2/genética , Interleucinas/metabolismo , Interleucinas/genética , Interleucina-18/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/imunologia , Camundongos Knockout , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Infecções por Salmonella/metabolismo , Caspase 1/metabolismo , Caspase 1/genética , Caspases Iniciadoras/metabolismo , Ceco/microbiologia , Colo/microbiologia , Colo/imunologia , Colo/metabolismo , Caspases/metabolismo , Citocinas/metabolismo , Transdução de Sinais , Mucinas/metabolismo , Salmonelose Animal/imunologia , Salmonelose Animal/microbiologia , Receptores de Superfície Celular
15.
Sci Rep ; 14(1): 22855, 2024 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-39353971

RESUMO

Periodontitis is a chronic inflammatory disease involving plaque biofilm as a pathogenic factor. Potassium ion plays an important role in cellular homeostasis; a large outflow of potassium may lead to local inflammation progression. In this work, the multifunctional short peptide molecule BmKTX-33 was designed by modifying the BmKTX, a Kv1.3 potassium channel inhibitor. This was to explore its antibacterial properties, capability of maintaining cell ion homeostasis, and bone-forming capacity. The results showed that BmKTX-33 had inhibitory effects on S. gordonii, F. nucleatum, and P. gingivalis. Moreover, BmKTX-33 also inhibited excessive potassium outflow in inflammatory environments. Finally, BmKTX-33 promoted MC3T3-E1 early osteogenesis while suppressing the NLRP3 inflammasome's production. In conclusion, BmKTX-33 not only has antibacterial properties, but also can inhibit the expression of NLRP3 inflammasome and play an anti-inflammatory role.


Assuntos
Proteína 3 que Contém Domínio de Pirina da Família NLR , Periodontite , Animais , Periodontite/tratamento farmacológico , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/uso terapêutico , Peptídeos/farmacologia , Peptídeos/química , Peptídeos/uso terapêutico , Porphyromonas gingivalis/efeitos dos fármacos , Potássio/metabolismo , Linhagem Celular , Osteogênese/efeitos dos fármacos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Anti-Inflamatórios/uso terapêutico , Biofilmes/efeitos dos fármacos
16.
Sci Rep ; 14(1): 24378, 2024 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-39420211

RESUMO

Systemic inflammation contributes to left ventricular (LV) dysfunction, however the role of the NLRP3 inflammasome in LV dysfunction in acute inflammatory conditions is unclear. This study investigated the role of the NLRP3 inflammasome in acute (24 h) cardiac structural and functional changes in vivo and in vitro in lipopolysaccharide (LPS)-induced inflammation. LPS-treated Sprague-Dawley (SD) rats showed increased LPS metabolite abundance in their LVs as measured by atmospheric pressure matrix-assisted laser desorption ionisation (AP-MALDI) mass spectrometry imaging (MSI). Echocardiography and histology showed that in LPS-exposed rats, LV internal diameter was decreased, with evidence of macrophage infiltration and oedema. However, there were no changes in LV wall thickness or collagen volume. Additionally, LPS-exposed rats exhibited impaired LV relaxation, potentially contributing to decreased stroke volume. While global systolic function was preserved, LPS exposure in SD rats resulted in impaired myocardial deformation assessed by speckle-tracking echocardiography. Exposure to LPS resulted in upregulation of the expression of components of the NLRP3 inflammasome in rodents. In vitro LPS exposure resulted in increased gene expression of NLRP3 and downstream cytokines IL-1ß and IL-18, antioxidant SOD2, and elevated markers of pyroptosis (GSDMD) which were inhibited by treatment with a NLRP3 antagonist. However, LPS-induced increases in the gene expression of apoptosic markers (BAX/Bcl2) were not impacted by NLRP3 antagonism. These findings suggest that inflammation induced adverse cardiac structural and functional changes is, at least in part, mediated by the NLRP3 inflammasome in acute, high-grade inflammatory states. In addition, in vitro findings suggest that while the NLRP3 inflammasome mediates pyroptotic pathways, regulation of apoptosis that is independent of the inflammasome.


Assuntos
Inflamassomos , Lipopolissacarídeos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ratos Sprague-Dawley , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Lipopolissacarídeos/efeitos adversos , Inflamassomos/metabolismo , Ratos , Masculino , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/patologia , Ecocardiografia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/induzido quimicamente , Piroptose/efeitos dos fármacos
17.
Nat Commun ; 15(1): 8595, 2024 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-39366977

RESUMO

The broadening of accessible methodologies has enabled mechanistic insights into single-pathogen infections, yet the molecular mechanisms underlying co-infections remain largely elusive, despite their clinical frequency and relevance, generally exacerbating symptom severity and fatality. Here, we describe an unbiased in vitro screening of pairwise co-infections in a murine macrophage model, quantifying pathogen proliferation and host cell death in parallel over time. The screen revealed that the majority of interactions are antagonistic for both metrics, highlighting general patterns depending on the pathogen virulence strategy. We subsequently decipher two distinct molecular interaction points: Firstly, murine Adenovirus 3 modifies ASC-dependent inflammasome responses in murine macrophages, altering host cell death and cytokine production, thereby impacting secondary Salmonella infection. Secondly, murine Adenovirus 2 infection triggers upregulation of Mprip, a crucial mediator of phagocytosis, which in turn causes increased Yersinia uptake, specifically in virus pre-infected bone-marrow-derived macrophages. This work therefore encompasses both a first-of-its-kind systematic assessment of host-pathogen-pathogen interactions, and mechanistic insight into molecular mediators during co-infection.


Assuntos
Coinfecção , Interações Hospedeiro-Patógeno , Inflamassomos , Macrófagos , Animais , Coinfecção/microbiologia , Coinfecção/virologia , Camundongos , Macrófagos/microbiologia , Macrófagos/virologia , Macrófagos/metabolismo , Inflamassomos/metabolismo , Sobrevivência Celular , Camundongos Endogâmicos C57BL , Fagocitose , Infecções por Adenoviridae/virologia , Citocinas/metabolismo , Infecções por Salmonella/microbiologia , Infecções por Salmonella/imunologia
18.
Sci Rep ; 14(1): 23816, 2024 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-39394435

RESUMO

Peritoneal inflammation and fibrosis remain major challenges to the long-term maintenance of peritoneal dialysis. Pemafibrate, a selective peroxisome proliferator-activated receptor α (PPARα) modulator, has been implicated in the management of fibrosis-related disorders. We investigated whether pemafibrate ameliorates peritoneal inflammation and fibrosis and explored the underlying mechanisms in mice with methylglyoxal (MGO)-induced peritoneal fibrosis (MGO mice). MGO mice exhibited peritoneal fibrosis with increased expression of mesenchymal markers, transforming growth factor-ß1 (TGF-ß1), and substantial deposition of extracellular matrix (ECM) proteins. Additionally, MGO mice exhibited peritoneal inflammation as indicated by elevated tumor necrosis factor-α expression and macrophage infiltration in peritoneal tissue. These effects were mitigated by pemafibrate treatment, which also restored peritoneal membrane function. Furthermore, pemafibrate promoted anti-inflammatory macrophage polarization in both mice and THP-1 cells. In human peritoneal mesothelial cells (HPMCs), pemafibrate effectively inhibited interferon-γ-induced production of TGF-ß1 and ECM while suppressing the proinflammatory cytokines nuclear factor-κB (NF-κB) and activator protein 1. The NF-κB inhibitory effect of pemafibrate involved stabilization of the NF-κB inhibitory protein IkBα. Notably, pemafibrate hindered activation of the NLR family pyrin domain containing 3/caspase-1 axis in interferon-γ-stimulated THP-1 cells. These findings suggest that pemafibrate ameliorates peritoneal inflammation and fibrosis, making it a promising candidate for peritoneal fibrosis therapy.


Assuntos
Benzoxazóis , Butiratos , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , PPAR alfa , Fibrose Peritoneal , Animais , PPAR alfa/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Camundongos , Humanos , Fibrose Peritoneal/tratamento farmacológico , Fibrose Peritoneal/metabolismo , Fibrose Peritoneal/patologia , Inflamassomos/metabolismo , Butiratos/farmacologia , Benzoxazóis/farmacologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Fator de Crescimento Transformador beta1/metabolismo , Masculino , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Peritonite/tratamento farmacológico , Peritonite/metabolismo , Peritonite/induzido quimicamente , Aldeído Pirúvico/metabolismo , Camundongos Endogâmicos C57BL , Células THP-1 , Modelos Animais de Doenças
19.
Cell Mol Biol Lett ; 29(1): 130, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39379817

RESUMO

Caspase-8, a renowned cysteine-aspartic protease within its enzyme family, initially garnered attention for its regulatory role in extrinsic apoptosis. With advancing research, a growing body of evidence has substantiated its involvement in other cell death processes, such as pyroptosis and necroptosis, as well as its modulatory effects on inflammasomes and proinflammatory cytokines. PANoptosis, an emerging concept of cell death, encompasses pyroptosis, apoptosis, and necroptosis, providing insight into the often overlapping cellular mortality observed during disease progression. The activation or deficiency of caspase-8 enzymatic activity is closely linked to PANoptosis, positioning caspase-8 as a key regulator of cell survival or death across various physiological and pathological processes. Aberrant expression of caspase-8 is closely associated with the development and progression of a range of inflammatory diseases, including immune system disorders, neurodegenerative diseases (NDDs), sepsis, and cancer. This paper delves into the regulatory role and impact of caspase-8 in these conditions, aiming to elucidate potential therapeutic strategies for the future intervention.


Assuntos
Caspase 8 , Inflamação , Caspase 8/metabolismo , Caspase 8/genética , Humanos , Animais , Piroptose , Necroptose/genética , Apoptose , Neoplasias/enzimologia , Inflamassomos/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/genética
20.
Front Cell Infect Microbiol ; 14: 1444178, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39463761

RESUMO

Introduction: Streptococcus uberis is a member of the pyogenic cluster of Streptococcus commonly associated with intramammary infection and mastitis in dairy cattle. It is a poorly controlled globally endemic pathogen responsible for a significant cause of the disease worldwide. The ruminant mammary gland provides an atypical body niche in which immune cell surveillance occurs on both sides of the epithelial tissue. S. uberis does not cause disease in non-ruminant species and is an asymptomatic commensal in other body niches. S. uberis exploits the unusual niche of the mammary gland to initiate an innate response from bovine mammary macrophage (BMMO) present in the secretion (milk) in which it can resist the host immune responses. As a result - and unexpectedly - the host inflammatory response is a key step in the pathogenesis of S.uberis, without which colonisation is impaired. In contrast to other bacteria pathogenic to the bovine mammary gland, S. uberis does not elicit innate responses from epithelial tissues; initial recognition of infection is via macrophages within milk. Methods: We dissected the role of the bacterial protein SUB1154 in the inflammasome pathway using ex vivo bovine mammary macrophages isolated from milk, recombinant protein expression, and a panel of inhibitors, agonists, and antagonists. We combine this with reverse-transcription quantitative real-time PCR to investigate the mechanisms underlying SUB1154-mediated priming of the immune response. Results: Here, we show that SUB1154 is responsible for priming the NLRP3 inflammasome in macrophages found in the mammary gland. Without SUB1154, IL-1ß is not produced, and we were able to restore IL-1ß responses to a sub1154 deletion S. uberis mutant using recombinant SUB1154. Surprisingly, only by blocking internalisation, or the cytoplasmic TIR domain of TLR2 were we able to block SUB1154-mediated priming. Discussion: Together, our data unifies several contrasting past studies and provides new mechanistic understanding of potential early interactions between pyogenic streptococci and the host.


Assuntos
Inflamassomos , Macrófagos , Glândulas Mamárias Animais , Infecções Estreptocócicas , Streptococcus , Receptor 2 Toll-Like , Animais , Bovinos , Inflamassomos/metabolismo , Inflamassomos/imunologia , Streptococcus/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 2 Toll-Like/genética , Feminino , Glândulas Mamárias Animais/microbiologia , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/metabolismo , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Mastite Bovina/microbiologia , Mastite Bovina/imunologia , Imunidade Inata , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Interleucina-1beta/metabolismo , Leite/microbiologia , Leite/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA