Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.838
Filtrar
1.
Immunol Lett ; 264: 36-45, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37940007

RESUMO

BACKGROUND: Intracerebral hemorrhage (ICH) is a serious medical problem, and promising strategy is limited. Macrophage initiated brain inflammatory injury following ICH, but the molecular mechanism had not been well identified. E3 ligase Nedd4L is implicated in the pathogenesis of the inflammatory immune response. METHODS: In the present study, we detected the levels of Nedd4L in macrophages following ICH. Furthermore, Macrophage M1 polarization, pro-inflammatory cytokine production, BBB disruption, brain water content and neurological function were examined in ICH mice. RESULTS: Here, we demonstrated that E3 ligase Nedd4L levels of macrophage increased following ICH, promoted M1 polarization inflammation by TRAF3. Nedd4L promoted BBB disruption, as well as neurological deficits. Inhibition of Nedd4L significantly attenuated M1 polarization in vivo. Inhibition of Nedd4L decreased TRAF3 and TBK1 levels, and subsequent phosphorylation of p38 and NF-κB p65 subunit following ICH. CONCLUSIONS: Our data demonstrated that Nedd4L was involved in the pathogenesis of ICH, which promoted inflammatory responses and exacerbated brain damage by TRAF3 following ICH.


Assuntos
Encéfalo , Hemorragia Cerebral , Ubiquitina-Proteína Ligases Nedd4 , Fator 3 Associado a Receptor de TNF , Animais , Camundongos , Encéfalo/imunologia , Encéfalo/patologia , Hemorragia Cerebral/imunologia , Hemorragia Cerebral/patologia , Macrófagos/enzimologia , Macrófagos/imunologia , Transdução de Sinais/fisiologia , Fator 3 Associado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligases Nedd4/metabolismo
2.
Phytochemistry ; 212: 113723, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37182686

RESUMO

A bioactivity-guided isolation from the aerial parts of Phyllanthus rheophyticus obtained 17 undescribed ent-cleistanthane-type diterpenoids, namely phyllarheophols A-Q, as well as 12 known analogs. Their structures were characterized by a combination of spectroscopic data interpretation, single-crystal X-ray diffraction and ECD analysis. The anti-inflammatory activities of these compounds were evaluated by measuring their inhibitory effects on NO production in LPS-stimulated RAW264.7 macrophages, and their preliminary structure-activity relationships were also discussed. Further study showed that promising compounds phyllarheophol D and phyacioid B significantly suppressed the expressions of cytokines and nitric oxide synthase through the NF-κB signaling pathway.


Assuntos
Anti-Inflamatórios , Diterpenos , Phyllanthus , Anti-Inflamatórios/química , Anti-Inflamatórios/isolamento & purificação , Anti-Inflamatórios/farmacologia , Diterpenos/química , Diterpenos/isolamento & purificação , Diterpenos/farmacologia , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/metabolismo , Estrutura Molecular , Óxido Nítrico/antagonistas & inibidores , Phyllanthus/química , Relação Estrutura-Atividade , NF-kappa B/metabolismo , Componentes Aéreos da Planta/química , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7 , Animais , Camundongos
3.
Nature ; 615(7952): 490-498, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36890227

RESUMO

Metabolic rewiring underlies the effector functions of macrophages1-3, but the mechanisms involved remain incompletely defined. Here, using unbiased metabolomics and stable isotope-assisted tracing, we show that an inflammatory aspartate-argininosuccinate shunt is induced following lipopolysaccharide stimulation. The shunt, supported by increased argininosuccinate synthase (ASS1) expression, also leads to increased cytosolic fumarate levels and fumarate-mediated protein succination. Pharmacological inhibition and genetic ablation of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) further increases intracellular fumarate levels. Mitochondrial respiration is also suppressed and mitochondrial membrane potential increased. RNA sequencing and proteomics analyses demonstrate that there are strong inflammatory effects resulting from FH inhibition. Notably, acute FH inhibition suppresses interleukin-10 expression, which leads to increased tumour necrosis factor secretion, an effect recapitulated by fumarate esters. Moreover, FH inhibition, but not fumarate esters, increases interferon-ß production through mechanisms that are driven by mitochondrial RNA (mtRNA) release and activation of the RNA sensors TLR7, RIG-I and MDA5. This effect is recapitulated endogenously when FH is suppressed following prolonged lipopolysaccharide stimulation. Furthermore, cells from patients with systemic lupus erythematosus also exhibit FH suppression, which indicates a potential pathogenic role for this process in human disease. We therefore identify a protective role for FH in maintaining appropriate macrophage cytokine and interferon responses.


Assuntos
Fumarato Hidratase , Interferon beta , Macrófagos , Mitocôndrias , RNA Mitocondrial , Humanos , Argininossuccinato Sintase/metabolismo , Ácido Argininossuccínico/metabolismo , Ácido Aspártico/metabolismo , Respiração Celular , Citosol/metabolismo , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/genética , Fumarato Hidratase/metabolismo , Fumaratos/metabolismo , Interferon beta/biossíntese , Interferon beta/imunologia , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Lúpus Eritematoso Sistêmico/enzimologia , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/metabolismo , Potencial da Membrana Mitocondrial , Metabolômica , Mitocôndrias/genética , Mitocôndrias/metabolismo , RNA Mitocondrial/metabolismo
4.
Int J Mol Sci ; 24(5)2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36902148

RESUMO

Chitotriosidase (CHIT1) is an enzyme produced by macrophages that regulates their differentiation and polarization. Lung macrophages have been implicated in asthma development; therefore, we asked whether pharmacological inhibition of macrophage-specific CHIT1 would have beneficial effects in asthma, as it has been shown previously in other lung disorders. CHIT1 expression was evaluated in the lung tissues of deceased individuals with severe, uncontrolled, steroid-naïve asthma. OATD-01, a chitinase inhibitor, was tested in a 7-week-long house dust mite (HDM) murine model of chronic asthma characterized by accumulation of CHIT1-expressing macrophages. CHIT1 is a dominant chitinase activated in fibrotic areas of the lungs of individuals with fatal asthma. OATD-01 given in a therapeutic treatment regimen inhibited both inflammatory and airway remodeling features of asthma in the HDM model. These changes were accompanied by a significant and dose-dependent decrease in chitinolytic activity in BAL fluid and plasma, confirming in vivo target engagement. Both IL-13 expression and TGFß1 levels in BAL fluid were decreased and a significant reduction in subepithelial airway fibrosis and airway wall thickness was observed. These results suggest that pharmacological chitinase inhibition offers protection against the development of fibrotic airway remodeling in severe asthma.


Assuntos
Remodelação das Vias Aéreas , Asma , Quitinases , Inibidores de Proteínas Quinases , Animais , Humanos , Camundongos , Remodelação das Vias Aéreas/efeitos dos fármacos , Asma/patologia , Asma/terapia , Quitinases/antagonistas & inibidores , Modelos Animais de Doenças , Pulmão/metabolismo , Macrófagos/enzimologia , Pyroglyphidae/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
5.
Nature ; 614(7948): 555-563, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36725935

RESUMO

Single-cell technologies have enabled the characterization of the tumour microenvironment at unprecedented depth and have revealed vast cellular diversity among tumour cells and their niche. Anti-tumour immunity relies on cell-cell relationships within the tumour microenvironment1,2, yet many single-cell studies lack spatial context and rely on dissociated tissues3. Here we applied imaging mass cytometry to characterize the immunological landscape of 139 high-grade glioma and 46 brain metastasis tumours from patients. Single-cell analysis of more than 1.1 million cells across 389 high-dimensional histopathology images enabled the spatial resolution of immune lineages and activation states, revealing differences in immune landscapes between primary tumours and brain metastases from diverse solid cancers. These analyses revealed cellular neighbourhoods associated with survival in patients with glioblastoma, which we leveraged to identify a unique population of myeloperoxidase (MPO)-positive macrophages associated with long-term survival. Our findings provide insight into the biology of primary and metastatic brain tumours, reinforcing the value of integrating spatial resolution to single-cell datasets to dissect the microenvironmental contexture of cancer.


Assuntos
Neoplasias Encefálicas , Glioma , Análise de Célula Única , Microambiente Tumoral , Humanos , Encéfalo/imunologia , Encéfalo/patologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Glioblastoma/imunologia , Glioblastoma/patologia , Glioma/imunologia , Glioma/patologia , Macrófagos/enzimologia , Microambiente Tumoral/imunologia , Metástase Neoplásica , Conjuntos de Dados como Assunto
6.
Nature ; 609(7927): 590-596, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36002575

RESUMO

Bacterial cell wall components provide various unique molecular structures that are detected by pattern recognition receptors (PRRs) of the innate immune system as non-self. Most bacterial species form a cell wall that consists of peptidoglycan (PGN), a polymeric structure comprising alternating amino sugars that form strands cross-linked by short peptides. Muramyl dipeptide (MDP) has been well documented as a minimal immunogenic component of peptidoglycan1-3. MDP is sensed by the cytosolic nucleotide-binding oligomerization domain-containing protein 24 (NOD2). Upon engagement, it triggers pro-inflammatory gene expression, and this functionality is of critical importance in maintaining a healthy intestinal barrier function5. Here, using a forward genetic screen to identify factors required for MDP detection, we identified N-acetylglucosamine kinase (NAGK) as being essential for the immunostimulatory activity of MDP. NAGK is broadly expressed in immune cells and has previously been described to contribute to the hexosamine biosynthetic salvage pathway6. Mechanistically, NAGK functions upstream of NOD2 by directly phosphorylating the N-acetylmuramic acid moiety of MDP at the hydroxyl group of its C6 position, yielding 6-O-phospho-MDP. NAGK-phosphorylated MDP-but not unmodified MDP-constitutes an agonist for NOD2. Macrophages from mice deficient in NAGK are completely deficient in MDP sensing. These results reveal a link between amino sugar metabolism and innate immunity to bacterial cell walls.


Assuntos
Acetilmuramil-Alanil-Isoglutamina , Proteína Adaptadora de Sinalização NOD2 , Fosfotransferases (Aceptor do Grupo Álcool) , Acetilmuramil-Alanil-Isoglutamina/química , Acetilmuramil-Alanil-Isoglutamina/imunologia , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Animais , Bactérias/química , Bactérias/imunologia , Parede Celular/química , Hexosaminas/biossíntese , Imunidade Inata , Macrófagos/enzimologia , Macrófagos/imunologia , Camundongos , Proteína Adaptadora de Sinalização NOD2/agonistas , Proteína Adaptadora de Sinalização NOD2/metabolismo , Peptidoglicano/química , Peptidoglicano/imunologia , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
7.
Cancer Lett ; 539: 215698, 2022 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-35523311

RESUMO

Macrophages play important roles in cancer microenvironment. Human cytosolic glycyl-tRNA synthetase (GARS1) was previously shown to be secreted via extracellular vesicles (EVs) from macrophages to trigger cancer cell death. However, the effects of GARS1-containing EVs (GARS1-EVs) on macrophages as well as on cancer cells and the working mechanisms of GARS1 in cancer microenvironment are not yet understood. Here we show that GARS1-EVs induce M1 polarization and facilitate phagocytosis of macrophages. GARS1-EVs triggers M1 polarization of macrophage via the specific interaction of the extracellular cadherin subdomains 1-4 of the cadherin EGF LAG seven-pass G-type receptor 2 (CELSR2) with the N-terminal WHEP domain containing peptide region of GARS1, and activates the RAF-MEK-ERK pathway for M1 type cytokine production and phagocytosis. Besides, GARS1 interacted with cadherin 6 (CDH6) of cancer cells via its C-terminal tRNA-binding domain to induce cancer cell death. In vivo model, GARS1-EVs showed potent suppressive activity against tumor initiation via M1 type macrophages. GARS1 displayed on macrophage-secreted extracellular vesicles suppressed tumor growth in dual mode, namely through pro-apoptotic effect on cancer cells and M1 polarization effect on macrophages. Collectively, these results elucidate the unique tumor suppressive activity and mechanism of GARS1-EVs by activating M1 macrophage via CELSR2 as well as by direct killing of cancer cells via CDH6.


Assuntos
Vesículas Extracelulares , Glicina-tRNA Ligase , Macrófagos , Neoplasias , Caderinas/metabolismo , Polaridade Celular , Vesículas Extracelulares/enzimologia , Vesículas Extracelulares/metabolismo , Glicina-tRNA Ligase/análise , Glicina-tRNA Ligase/metabolismo , Glicina-tRNA Ligase/farmacologia , Humanos , Macrófagos/enzimologia , Macrófagos/metabolismo , Macrófagos/patologia , Neoplasias/enzimologia , Neoplasias/metabolismo , Fagocitose , Microambiente Tumoral
8.
J Biol Chem ; 298(5): 101857, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35337798

RESUMO

Innate immune signaling by Toll-like receptors (TLRs) involves receptor phosphorylation, which helps to shape and drive key inflammatory outputs, yet our understanding of the kinases and mechanisms that mediate TLR phosphorylation is incomplete. Spleen tyrosine kinase (Syk) is a nonreceptor protein tyrosine kinase, which is known to relay adaptive and innate immune signaling, including from TLRs. However, TLRs do not contain the conserved dual immunoreceptor tyrosine-based activation motifs that typically recruit Syk to many other receptors. One possibility is that the Syk-TLR association is indirect, relying on an intermediary scaffolding protein. We previously identified a role for the palmitoylated transmembrane adapter protein SCIMP in scaffolding the Src tyrosine kinase Lyn, for TLR phosphorylation, but the role of SCIMP in mediating the interaction between Syk and TLRs has not yet been investigated. Here, we show that SCIMP recruits Syk in response to lipopolysaccharide-mediated TLR4 activation. We also show that Syk contributes to the phosphorylation of SCIMP and TLR4 to enhance their binding. Further evidence pinpoints two specific phosphorylation sites in SCIMP critical for its interaction with Syk-SH2 domains in the absence of immunoreceptor tyrosine-based activation motifs. Finally, using inhibitors and primary macrophages from SCIMP-/- mice, we confirm a functional role for SCIMP-mediated Syk interaction in modulating TLR4 phosphorylation, signaling, and cytokine outputs. In conclusion, we identify SCIMP as a novel, immune-specific Syk scaffold, which can contribute to inflammation through selective TLR-driven inflammatory responses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Membrana , Quinase Syk , Receptor 4 Toll-Like , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Macrófagos/enzimologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Quinase Syk/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Tirosina/metabolismo , Quinases da Família src/metabolismo
9.
Cells ; 11(3)2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35159206

RESUMO

Sterile inflammation either resolves the initial insult or leads to tissue damage. Kidney ischemia/reperfusion injury (IRI) is associated with neutrophilic infiltration, enhanced production of inflammatory mediators, accumulation of necrotic cells and tissue remodeling. Macrophage-dependent microenvironmental changes orchestrate many features of the immune response and tissue regeneration. The activation status of macrophages is influenced by extracellular signals, the duration and intensity of the stimulation, as well as various regulatory molecules. The role of macrophage-derived monocyte chemoattractant protein-induced protein 1 (MCPIP1), also known as Regnase-1, in kidney ischemia-reperfusion injury (IRI) and recovery from sterile inflammation remains unresolved. In this study, we showed that macrophage-specific Mcpip1 deletion significantly affects the kidney phenotype. Macrophage-specific Mcpip1 transgenic mice displayed enhanced inflammation and loss of the tubular compartment upon IRI. We showed that MCPIP1 modulates sterile inflammation by negative regulation of Irf4 expression and accumulation of IRF4+ cells in the tissue and, consequently, suppresses the post-ischemic kidney immune response. Thus, we identified MCPIP1 as an important molecular sentinel of immune homeostasis in experimental acute kidney injury (AKI) and renal fibrosis.


Assuntos
Injúria Renal Aguda , Rim , Traumatismo por Reperfusão , Ribonucleases/genética , Injúria Renal Aguda/metabolismo , Animais , Inflamação/metabolismo , Rim/metabolismo , Rim/patologia , Macrófagos/enzimologia , Camundongos , Proteínas Quimioatraentes de Monócitos/metabolismo , Traumatismo por Reperfusão/metabolismo
10.
Clin Sci (Lond) ; 136(4): 273-289, 2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35132999

RESUMO

BACKGROUND: NOD-like receptor family pyrin domain containing 3 (NLRP3)-mediated macrophage pyroptosis plays an important role in sepsis-induced acute lung injury (ALI). Inhibition of pyroptosis may be a way to alleviate inflammation as well as tissue damage triggered after lipopolysaccharide (LPS) stimulation. The aim of the present study was to explore whether buformin (BF), a hypoglycemic agent, could alleviate sepsis-induced ALI by inhibiting pyroptosis. METHODS: Wildtype C57BL/6 mice were randomly divided into control group, BF group, LPS group and LPS+BF group. BF group and LPS+BF group were pretreated with BF at a dose of 25 mg/kg, and the changes were observed. In addition, BF was used to interfere with THP-1 cells. The therapeutic effect of BF has been verified by intraperitoneal injection of BF in vivo after LPS stimulation. RESULTS: Inflammation and injury was significantly reduced in BF pretreated mice, and the indexes related to pyroptosis were suppressed. The phosphorylation of AMP-activated protein kinase (AMPK) in lung tissues of mice in the BF and LPS+BF groups was significantly higher. In THP-1 cells, the AMPK inhibitor, Compound C was added to demonstrate that BF worked via AMPK to inhibit NLRP3 inflammasome. It was further demonstrated that BF up-regulated autophagy, which in turn promoted NLRP3 inflammasome degradation. On the other hand, BF decreased NLRP3 mRNA level by increasing nuclear factor-erythroid 2 related factor 2 (Nrf2). And BF showed a therapeutic effect after LPS challenge. CONCLUSION: Our study confirmed that BF inhibited NLRP3-mediated pyroptosis in sepsis-induced ALI by up-regulating autophagy and Nrf2 protein level through an AMPK-dependent pathway. This provides a new strategy for clinical mitigation of sepsis-induced ALI.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Buformina/uso terapêutico , Hipoglicemiantes/uso terapêutico , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Animais , Autofagia/efeitos dos fármacos , Buformina/farmacologia , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Humanos , Hipoglicemiantes/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/metabolismo , Sepse/complicações
11.
Sci Rep ; 12(1): 1383, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-35082324

RESUMO

Bacillus subtilis, a probiotic, has been applied in the medical, food, and feed industries among others. However, the mechanisms of its benefits to hosts are not yet fully understood. Here the characterization and bioactivities of an extracellular polymeric substance (EPS) from Bacillus subtilis were investigated to reveal its partial mechanisms and provide the theoretical basics for further development and utilization of Bacillus subtilis. In this study, the novel strain Bacillus subtilis xztubd1 (GenBank: MG458322.1) was isolated from a housefly's body, identified according to phenotypical and genotypical analyses, and found to produce large amounts of an EPS. Through ultraviolet spectroscopy and Fourier transform infrared spectroscopy (FTIR spectroscopy), the EPS was found to contain a variety of chemical functional groups, such as O-H groups, C=C, C=O, CH3, C-O-H and C-O-C bonds, and alpha-type pyranose. Furthermore, the in vitro antioxidant activity of the EPS on DPPH radicals at a concentration of 90 µg/ml was 62%; on the superoxide radical at a concentration of 90 µg/ml, this value was 75%; and on hydroxyl radicals at a concentration of 90 µg/ml, the activity was 54%. EPS also enhanced significantly phagocytosis, lysozyme activity in macrophages, IL-2 content in mice and inhibited dramatically the growth of HeLa cells. These results showed that the EPS with reductive groups have the strong capacity to scavenge reactive oxygen species (ROS), reinforce the immune system and inhibit the growth of cancer cell, which helps theirs hosts defence against many diseases, including inflammation and cancer. The EPS from Bacillus subtilis has the potential to be an anticancer and anti-inflammatory drug candidate in the pharmaceutical industries, which provide scientific evidence for the development and utilization of probiotic-derived medicines.


Assuntos
Anti-Inflamatórios/administração & dosagem , Antineoplásicos/administração & dosagem , Bacillus subtilis/isolamento & purificação , Bacillus subtilis/metabolismo , Moscas Domésticas/microbiologia , Polissacarídeos Bacterianos/administração & dosagem , Probióticos/administração & dosagem , Animais , Animais não Endogâmicos , Anti-Inflamatórios/química , Anti-Inflamatórios/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Antioxidantes/administração & dosagem , Antioxidantes/química , Antioxidantes/metabolismo , Proliferação de Células/efeitos dos fármacos , Matriz Extracelular de Substâncias Poliméricas/química , Matriz Extracelular de Substâncias Poliméricas/metabolismo , Células HeLa , Humanos , Interleucina-2/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Masculino , Camundongos , Muramidase/metabolismo , Fagocitose/efeitos dos fármacos , Polissacarídeos Bacterianos/biossíntese , Probióticos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Espectroscopia de Infravermelho com Transformada de Fourier/métodos
12.
Mol Cell ; 82(3): 527-541.e7, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35016033

RESUMO

Citrulline can be converted into argininosuccinate by argininosuccinate synthetase (ASS1) in the urea cycle and the citrulline-nitric oxide cycle. However, the regulation and biological function of citrulline metabolism remain obscure in the immune system. Unexpectedly, we found that macrophage citrulline declines rapidly after interferon gamma (IFN-γ) and/or lipopolysaccharide (LPS) stimulation, which is required for efficient proinflammatory signaling activation. Mechanistically, IFN-γ and/or LPS stimulation promotes signal transducers and activators of transcription 1 (STAT1)-mediated ASS1 transcription and Janus kinase2 (JAK2)-mediated phosphorylation of ASS1 at tyrosine 87, thereby leading to citrulline depletion. Reciprocally, increased citrulline directly binds to JAK2 and inhibits JAK2-STAT1 signaling. Blockage of ASS1-mediated citrulline depletion suppresses the host defense against bacterial infection in vivo. We therefore define a central role for ASS1 in controlling inflammatory macrophage activation and antibacterial defense through depletion of cellular citrulline and, further, identify citrulline as an innate immune-signaling metabolite that engages a metabolic checkpoint for proinflammatory responses.


Assuntos
Argininossuccinato Sintase/metabolismo , Citrulina/metabolismo , Imunidade Inata , Inflamação/enzimologia , Listeriose/enzimologia , Ativação de Macrófagos , Macrófagos/enzimologia , Animais , Argininossuccinato Sintase/genética , Modelos Animais de Doenças , Células HEK293 , Humanos , Inflamação/genética , Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Listeria monocytogenes/imunologia , Listeriose/genética , Listeriose/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Células RAW 264.7 , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
13.
Am J Physiol Renal Physiol ; 322(1): F105-F119, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34866403

RESUMO

15-Lipoxygenase (15-LO) is a nonheme iron-containing dioxygenase that has both pro- and anti-inflammatory roles in many tissues and disease states. 15-LO is thought to influence macrophage phenotype, and silencing 15-LO reduces fibrosis after acute inflammatory triggers. The goal of the present study was to determine whether altering 15-LO expression influences inflammation and fibrogenesis in a murine model of unilateral ureteral obstruction (UUO). C57BL/6J mice, 15-LO knockout (Alox15-/-) mice, and 15-LO transgenic overexpressing (15LOTG) mice were subjected UUO, and kidneys were analyzed at 3, 10, and 14 days postinjury. Histology for fibrosis, inflammation, cytokine quantification, flow cytometry, and metabolomics were performed on injured tissues and controls. PD146176, a specific 15-LO inhibitor, was used to complement experiments involving knockout animals. Compared with wild-type animals undergoing UUO, Alox15-/- mouse kidneys had less proinflammatory, profibrotic message along with less fibrosis and macrophage infiltration. PD146176 inhibited 15-LO and resulted in reduced fibrosis and macrophage infiltration similar to Alox15-/- mice. Flow cytometry revealed that Alox15-/- UUO-injured kidneys had a dynamic change in macrophage phenotype, with an early blunting of CD11bHiLy6CHi "M1" macrophages and an increase in anti-inflammatory CD11bHiLy6CInt "M2c" macrophages and reduced expression of the fractalkine receptor chemokine (C-X3-C motif) receptor 1. Many of these findings were reversed when UUO was performed on 15LOTG mice. Metabolomics analysis revealed that wild-type kidneys developed a glycolytic shift postinjury, while Alox15-/- kidneys exhibited increased oxidative phosphorylation. In conclusion, 15-LO manipulation by genetic or pharmacological means induces dynamic changes in the inflammatory microenvironment in the UUO model and appears to be critical in the progression of UUO-induced fibrosis.NEW & NOTEWORTHY 15-Lipoxygenase (15-LO) has both pro- and anti-inflammatory functions in leukocytes, and its role in kidney injury and repair is unexplored. Our study showed that 15-LO worsens inflammation and fibrosis in a rodent model of chronic kidney disease using genetic and pharmacological manipulation. Silencing 15-LO promotes an increase in M2c-like wound-healing macrophages in the kidney and alters kidney metabolism globally, protecting against anaerobic glycolysis after injury.


Assuntos
Araquidonato 12-Lipoxigenase/metabolismo , Araquidonato 15-Lipoxigenase/metabolismo , Citocinas/metabolismo , Metabolismo Energético , Mediadores da Inflamação/metabolismo , Rim/enzimologia , Metaboloma , Nefrite/etiologia , Obstrução Ureteral/complicações , Animais , Araquidonato 12-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/genética , Microambiente Celular , Citocinas/genética , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Fibrose , Rim/efeitos dos fármacos , Rim/patologia , Leucócitos/enzimologia , Inibidores de Lipoxigenase/farmacologia , Macrófagos/enzimologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrite/enzimologia , Nefrite/patologia , Nefrite/prevenção & controle , Fenótipo , Obstrução Ureteral/tratamento farmacológico , Obstrução Ureteral/enzimologia , Obstrução Ureteral/patologia
14.
J Biol Chem ; 298(2): 101501, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34929172

RESUMO

Activated macrophages undergo metabolic reprogramming, which not only supports their energetic demands but also allows for the production of specific metabolites that function as signaling molecules. Several Krebs cycles, or Krebs-cycle-derived metabolites, including succinate, α-ketoglutarate, and itaconate, have recently been shown to modulate macrophage function. The accumulation of 2-hydroxyglutarate (2HG) has also been well documented in transformed cells and more recently shown to play a role in T cell and dendritic cell function. Here we have found that the abundance of both enantiomers of 2HG is increased in LPS-activated macrophages. We show that L-2HG, but not D-2HG, can promote the expression of the proinflammatory cytokine IL-1ß and the adoption of an inflammatory, highly glycolytic metabolic state. These changes are likely mediated through activation of the transcription factor hypoxia-inducible factor-1α (HIF-1α) by L-2HG, a known inhibitor of the HIF prolyl hydroxylases. Expression of the enzyme responsible for L-2HG degradation, L-2HG dehydrogenase (L-2HGDH), was also found to be decreased in LPS-stimulated macrophages and may therefore also contribute to L-2HG accumulation. Finally, overexpression of L-2HGDH in HEK293 TLR4/MD2/CD14 cells inhibited HIF-1α activation by LPS, while knockdown of L-2HGDH in macrophages boosted the induction of HIF-1α-dependent genes, as well as increasing LPS-induced HIF-1α activity. Taken together, this study therefore identifies L-2HG as a metabolite that can regulate HIF-1α in macrophages.


Assuntos
Glutaratos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Lipopolissacarídeos , Macrófagos , Glutaratos/metabolismo , Células HEK293 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/metabolismo
15.
Front Immunol ; 12: 779235, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925356

RESUMO

The host immune system plays a pivotal role in the containment of Mycobacterium tuberculosis (Mtb) infection, and host-directed therapy (HDT) is emerging as an effective strategy to treat tuberculosis (TB), especially drug-resistant TB. Previous studies revealed that expression of sirtuin 7 (SIRT7), a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase, was downregulated in macrophages after Mycobacterial infection. Inhibition of SIRT7 with the pan-sirtuin family inhibitor nicotinamide (NAM), or by silencing SIRT7 expression, promoted intracellular growth of Mtb and restricted the generation of nitric oxide (NO). Addition of the exogenous NO donor SNAP abrogated the increased bacterial burden in NAM-treated or SIRT7-silenced macrophages. Furthermore, SIRT7-silenced macrophages displayed a lower frequency of early apoptotic cells after Mycobacterial infection, and this could be reversed by providing exogenous NO. Overall, this study clarified a SIRT7-mediated protective mechanism against Mycobacterial infection through regulation of NO production and apoptosis. SIRT7 therefore has potential to be exploited as a novel effective target for HDT of TB.


Assuntos
Apoptose , Macrófagos/enzimologia , Mycobacterium tuberculosis/imunologia , Óxido Nítrico/metabolismo , Fagocitose , Sirtuínas/metabolismo , Tuberculose/enzimologia , Animais , Antituberculosos/farmacologia , Apoptose/efeitos dos fármacos , Carga Bacteriana , Interações Hospedeiro-Patógeno , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/patogenicidade , Doadores de Óxido Nítrico/farmacologia , Células RAW 264.7 , S-Nitroso-N-Acetilpenicilamina/farmacologia , Transdução de Sinais , Sirtuínas/genética , Tuberculose/tratamento farmacológico , Tuberculose/imunologia , Tuberculose/microbiologia
16.
Front Immunol ; 12: 748573, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34759927

RESUMO

The cytokine storm is a marker of severity of various diseases and increased mortality. The altered metabolic profile and energy generation of immune cells affects their activation, exacerbating the cytokine storm. Currently, the emerging field of immunometabolism has highlighted the importance of specific metabolic pathways in immune regulation. The glycolytic enzyme pyruvate kinase M2 (PKM2) is a key regulator of immunometabolism and bridges metabolic and inflammatory dysfunction. This enzyme changes its conformation thus walks in different fields including metabolism and inflammation and associates with various transcription factors. This review summarizes the vital role of PKM2 in mediating immunometabolic reprogramming and its role in inducing cytokine storm, with a focus on providing references for further understanding of its pathological functions and for proposing new targets for the treatment of related diseases.


Assuntos
Proteínas de Transporte/fisiologia , Síndrome da Liberação de Citocina/enzimologia , Proteínas de Membrana/fisiologia , Hormônios Tireóideos/fisiologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proteínas de Transporte/agonistas , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/química , Núcleo Celular/enzimologia , Citocinas/metabolismo , Células Dendríticas/enzimologia , Ativação Enzimática , Ferroptose , Glicólise , Humanos , Inflamassomos , Inflamação , Células Matadoras Naturais/enzimologia , Macrófagos/enzimologia , Proteínas de Membrana/agonistas , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/química , Estrutura Molecular , Oxirredução , Conformação Proteica , Piroptose , Hormônios Tireóideos/agonistas , Hormônios Tireóideos/química , Proteínas de Ligação a Hormônio da Tireoide
17.
Cells ; 10(11)2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34831416

RESUMO

Nitric oxide and its production by iNOS is an established mechanism critical to tumor promotion or suppression. Macrophages have important roles in immunity, development, and progression of cancer and have a controversial role in pro- and antitumoral effects. The tumor microenvironment consists of tumor-associated macrophages (TAM), among other cell types that influence the fate of the growing tumor. Depending on the microenvironment and various cues, macrophages polarize into a continuum represented by the M1-like pro-inflammatory phenotype or the anti-inflammatory M2-like phenotype; these two are predominant, while there are subsets and intermediates. Manipulating their plasticity through programming or reprogramming of M2-like to M1-like phenotypes presents the opportunity to maximize tumoricidal defenses. The dual role of iNOS-derived NO also influences TAM activity by repolarization to tumoricidal M1-type phenotype. Regulatory pathways and immunomodulation achieve this through miRNA that may inhibit the immunosuppressive tumor microenvironment. This review summarizes the classical physiology of macrophages and polarization, iNOS activities, and evidence towards TAM reprogramming with current information in glioblastoma and melanoma models, and the immunomodulatory and therapeutic options using iNOS or NO-dependent strategies.


Assuntos
Reprogramação Celular , Macrófagos/enzimologia , Macrófagos/patologia , Neoplasias/patologia , Neoplasias/terapia , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Animais , Plasticidade Celular , Humanos
18.
Sci Rep ; 11(1): 22901, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34824340

RESUMO

Macrophages serve as viral reservoirs due to their resistance to apoptosis and HIV-cytopathic effects. We have previously shown that inhibitor of apoptosis proteins (IAPs) confer resistance to HIV-Vpr-induced apoptosis in normal macrophages. Herein, we show that second mitochondrial activator of caspases (SMAC) mimetics (SM) induce apoptosis of monocyte-derived macrophages (MDMs) infected in vitro with a R5-tropic laboratory strain expressing heat stable antigen, chronically infected U1 cells, and ex-vivo derived MDMs from HIV-infected individuals. To understand the mechanism governing SM-induced cell death, we show that SM-induced cell death of primary HIV-infected macrophages was independent of the acquisition of M1 phenotype following HIV infection of macrophages. Instead, SM-induced cell death was found to be mediated by IAPs as downregulation of IAPs by siRNAs induced cell death of HIV-infected macrophages. Moreover, HIV infection caused receptor interacting protein kinase-1 (RIPK1) degradation which in concert with IAP1/2 downregulation following SM treatment may result in apoptosis of macrophages. Altogether, our results show that SM selectively induce apoptosis in primary human macrophages infected in vitro with HIV possibly through RIPK1. Moreover, modulation of the IAP pathways may be a potential strategy for selective killing of HIV-infected macrophages in vivo.


Assuntos
Fármacos Anti-HIV/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , HIV-1/patogenicidade , Macrófagos/efeitos dos fármacos , Mimetismo Molecular , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteína 3 com Repetições IAP de Baculovírus/genética , Proteína 3 com Repetições IAP de Baculovírus/metabolismo , Efeito Citopatogênico Viral , Infecções por HIV/enzimologia , Infecções por HIV/patologia , Infecções por HIV/virologia , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Macrófagos/enzimologia , Macrófagos/patologia , Macrófagos/virologia , Fenótipo , Células U937 , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
19.
mBio ; 12(6): e0281721, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34844429

RESUMO

HIV-1 replicates in cells that express a wide array of innate immune sensors and may do so simultaneously with other pathogens. How a coexisting innate immune stimulus influences the outcome of HIV-1 sensing, however, remains poorly understood. Here, we demonstrate that the activation of a second signaling pathway enables a cyclic GMP-AMP synthase (cGAS)-dependent type I interferon (IFN-I) response to HIV-1 infection. We used RNA sequencing to determine that HIV-1 alone induced few or no signs of an IFN-I response in THP-1 cells. In contrast, when supplemented with suboptimal levels of bacterial lipopolysaccharide (LPS), HIV-1 infection triggered the production of elevated levels of IFN-I and significant upregulation of interferon-stimulated genes. LPS-mediated enhancement of IFN-I production upon HIV-1 infection, which was observed in primary macrophages, was lost by blocking reverse transcription and with a hyperstable capsid, pointing to viral DNA being an essential immunostimulatory molecule. LPS also synergistically enhanced IFN-I production by cyclic GMP-AMP (cGAMP), a second messenger of cGAS. These observations suggest that the DNA sensor cGAS is responsible for a type I IFN response to HIV-1 in concert with LPS receptor Toll-like receptor 4 (TLR4). Small amounts of a TLR2 agonist also cooperate with HIV-1 to induce type I IFN production. These results demonstrate how subtle immunomodulatory activity renders HIV-1 capable of eliciting an IFN-I response through positive cross talk between cGAS and TLR sensing pathways. IMPORTANCE Innate immune activation is a hallmark of HIV-1 pathogenesis. Thus, it is critical to understand how HIV-1 infection elicits innate immune responses. In this work, we show that HIV-1 infection of macrophages leads to a robust type I interferon (IFN) production only when a second signaling event is initiated by a coexisting immunostimulatory molecule. Our results show that HIV-1 infection alone is not sufficient for triggering a strong IFN response. We find that bacterial membrane components, which are recognized by endosomal innate sensors, enable production of elevated levels of IFNs and significant upregulation of interferon-stimulated genes upon HIV-1 infection. This IFN response is dependent on viral DNA synthesis and prevented by a stable capsid, pointing to an essential role for a DNA sensing molecule. These observations provide new insights into how different innate immune recognition pathways synergize during HIV-1 infection and determine the outcome of innate responses.


Assuntos
Infecções por HIV/imunologia , HIV-1/fisiologia , Macrófagos/virologia , Nucleotidiltransferases/imunologia , Receptor 4 Toll-Like/imunologia , Infecções por HIV/enzimologia , Infecções por HIV/genética , Infecções por HIV/virologia , Humanos , Imunidade Inata , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Macrófagos/enzimologia , Macrófagos/imunologia , Nucleotídeos Cíclicos , Nucleotidiltransferases/genética , Transdução de Sinais , Receptor 4 Toll-Like/genética
20.
Exp Eye Res ; 213: 108823, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34752817

RESUMO

Choroidal neovascularization (CNV), a feature of neovasular age-related macular degeneration (AMD), acts as a leading cause of vision loss in the elderly. Shikonin (SHI), a natural bioactive compound extracted from Chinese herb radix arnebiae, exerts anti-inflammatory and anti-angiogenic roles and also acts as a potential pyruvate kinase M2 (PKM2) inhibitor in macrophages. The major immune cells macrophages infiltrate the CNV lesions, where the production of pro-angiognic cytokines from macrophage facilitates the development of CNV. PKM2 contributes to the neovascular diseases. In this study, we found that SHI oral gavage alleviated the leakage, area and volume of mouse laser-induced CNV lesion and inhibited macrophage infiltration without ocular cytotoxicity. Moreover, SHI inhibited the secretion of pro-angiogenic cytokine, including basic fibroblast growth factor (FGF2), insulin-like growth factor-1 (IGF1), chemokine (C-C motif) ligand 2 (CCL2), placental growth factor and vascular endothelial growth factor (VEGF), from primary human macrophages by down-regulating PKM2/STAT3/CD163 pathway, indicating a novel potential therapy strategy for CNV.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anti-Inflamatórios não Esteroides/uso terapêutico , Neovascularização de Coroide/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Naftoquinonas/uso terapêutico , Piruvato Quinase/antagonistas & inibidores , Indutores da Angiogênese/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Western Blotting , Células Cultivadas , Neovascularização de Coroide/enzimologia , Cromatografia Líquida de Alta Pressão , Corantes/administração & dosagem , Citocinas/metabolismo , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/uso terapêutico , Ensaio de Imunoadsorção Enzimática , Angiofluoresceinografia , Humanos , Marcação In Situ das Extremidades Cortadas , Verde de Indocianina/administração & dosagem , Macrófagos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Piruvato Quinase/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA