Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.172
Filtrar
1.
PLoS Pathog ; 20(5): e1012148, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38728367

RESUMO

Previously, we found that Mycobacterium tuberculosis (Mtb) infection in type 2 diabetes mellitus (T2DM) mice enhances inflammatory cytokine production which drives pathological immune responses and mortality. In the current study, using a T2DM Mtb infection mice model, we determined the mechanisms that make T2DM mice alveolar macrophages (AMs) more inflammatory upon Mtb infection. Among various cell death pathways, necroptosis is a major pathway involved in inflammatory cytokine production by T2DM mice AMs. Anti-TNFR1 antibody treatment of Mtb-infected AMs from T2DM mice significantly reduced expression of receptor interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL) (necroptosis markers) and IL-6 production. Metabolic profile comparison of Mtb-infected AMs from T2DM mice and Mtb-infected AMs of nondiabetic control mice indicated that 2-ketohexanoic acid and deoxyadenosine monophosphate were significantly abundant, and acetylcholine and pyridoxine (Vitamin B6) were significantly less abundant in T2DM mice AMs infected with Mtb. 2-Ketohexanoic acid enhanced expression of TNFR1, RIPK3, MLKL and inflammatory cytokine production in the lungs of Mtb-infected nondiabetic mice. In contrast, pyridoxine inhibited RIPK3, MLKL and enhanced expression of Caspase 3 (apoptosis marker) in the lungs of Mtb-infected T2DM mice. Our findings demonstrate that metabolic changes in Mtb-infected T2DM mice enhance TNFR1-mediated necroptosis of AMs, which leads to excess inflammation and lung pathology.


Assuntos
Diabetes Mellitus Tipo 2 , Mycobacterium tuberculosis , Necroptose , Animais , Camundongos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/microbiologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Camundongos Endogâmicos C57BL , Tuberculose/imunologia , Tuberculose/metabolismo , Tuberculose/microbiologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/microbiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Masculino , Citocinas/metabolismo
2.
Front Immunol ; 15: 1380628, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38774866

RESUMO

Introduction: TAM receptor-mediated efferocytosis plays an important function in immune regulation and may contribute to antigen tolerance in the lungs, a site with continuous cellular turnover and generation of apoptotic cells. Some studies have identified failures in efferocytosis as a common driver of inflammation and tissue destruction in lung diseases. Our study is the first to characterize the in vivo function of the TAM receptors, Axl and MerTk, in the innate immune cell compartment, cytokine and chemokine production, as well as the alveolar macrophage (AM) phenotype in different settings in the airways and lung parenchyma. Methods: We employed MerTk and Axl defective mice to induce acute silicosis by a single exposure to crystalline silica particles (20 mg/50 µL). Although both mRNA levels of Axl and MerTk receptors were constitutively expressed by lung cells and isolated AMs, we found that MerTk was critical for maintaining lung homeostasis, whereas Axl played a role in the regulation of silica-induced inflammation. Our findings imply that MerTk and Axl differently modulated inflammatory tone via AM and neutrophil recruitment, phenotype and function by flow cytometry, and TGF-ß and CXCL1 protein levels, respectively. Finally, Axl expression was upregulated in both MerTk-/- and WT AMs, confirming its importance during inflammation. Conclusion: This study provides strong evidence that MerTk and Axl are specialized to orchestrate apoptotic cell clearance across different circumstances and may have important implications for the understanding of pulmonary inflammatory disorders as well as for the development of new approaches to therapy.


Assuntos
Receptor Tirosina Quinase Axl , Homeostase , Pulmão , Macrófagos Alveolares , Camundongos Knockout , Proteínas Proto-Oncogênicas , Receptores Proteína Tirosina Quinases , Silicose , c-Mer Tirosina Quinase , Animais , c-Mer Tirosina Quinase/metabolismo , c-Mer Tirosina Quinase/genética , Silicose/metabolismo , Silicose/imunologia , Silicose/patologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases/genética , Camundongos , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos Endogâmicos C57BL , Citocinas/metabolismo , Modelos Animais de Doenças
3.
Front Immunol ; 15: 1372959, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38690277

RESUMO

Introduction: Hypoxia is a common pathological driver contributing to various forms of pulmonary vascular diseases leading to pulmonary hypertension (PH). Pulmonary interstitial macrophages (IMs) play pivotal roles in immune and vascular dysfunction, leading to inflammation, abnormal remodeling, and fibrosis in PH. However, IMs' response to hypoxia and their role in PH progression remain largely unknown. We utilized a murine model of hypoxia-induced PH to investigate the repertoire and functional profiles of IMs in response to acute and prolonged hypoxia, aiming to elucidate their contributions to PH development. Methods: We conducted single-cell transcriptomic analyses to characterize the repertoire and functional profiles of murine pulmonary IMs following exposure to hypobaric hypoxia for varying durations (0, 1, 3, 7, and 21 days). Hallmark pathways from the mouse Molecular Signatures Database were utilized to characterize the molecular function of the IM subpopulation in response to hypoxia. Results: Our analysis revealed an early acute inflammatory phase during acute hypoxia exposure (Days 1-3), which was resolved by Day 7, followed by a pro-remodeling phase during prolonged hypoxia (Days 7-21). These phases were marked by distinct subpopulations of IMs: MHCIIhiCCR2+EAR2+ cells characterized the acute inflammatory phase, while TLF+VCAM1hi cells dominated the pro-remodeling phase. The acute inflammatory phase exhibited enrichment in interferon-gamma, IL-2, and IL-6 pathways, while the pro-remodeling phase showed dysregulated chemokine production, hemoglobin clearance, and tissue repair profiles, along with activation of distinct complement pathways. Discussion: Our findings demonstrate the existence of distinct populations of pulmonary interstitial macrophages corresponding to acute and prolonged hypoxia exposure, pivotal in regulating the inflammatory and remodeling phases of PH pathogenesis. This understanding offers potential avenues for targeted interventions, tailored to specific populations and distinct phases of the disease. Moreover, further identification of triggers for pro-remodeling IMs holds promise in unveiling novel therapeutic strategies for pulmonary hypertension.


Assuntos
Perfilação da Expressão Gênica , Hipertensão Pulmonar , Hipóxia , Análise de Célula Única , Transcriptoma , Animais , Camundongos , Hipóxia/metabolismo , Hipóxia/imunologia , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/imunologia , Hipertensão Pulmonar/genética , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Masculino , Pulmão/imunologia , Pulmão/patologia , Pulmão/metabolismo
4.
Nat Commun ; 15(1): 4326, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773113

RESUMO

Resolving inflammation is thought to return the affected tissue back to homoeostasis but recent evidence supports a non-linear model of resolution involving a phase of prolonged immune activity. Here we show that within days following resolution of Streptococcus pneumoniae-triggered lung inflammation, there is an influx of antigen specific lymphocytes with a memory and tissue-resident phenotype as well as macrophages bearing alveolar or interstitial phenotype. The transcriptome of these macrophages shows enrichment of genes associated with prostaglandin biosynthesis and genes that drive T cell chemotaxis and differentiation. Therapeutic depletion of post-resolution macrophages, inhibition of prostaglandin E2 (PGE2) synthesis or treatment with an EP4 antagonist, MF498, reduce numbers of lung CD4+/CD44+/CD62L+ and CD4+/CD44+/CD62L-/CD27+ T cells as well as their expression of the α-integrin, CD103. The T cells fail to reappear and reactivate upon secondary challenge for up to six weeks following primary infection. Concomitantly, EP4 antagonism through MF498 causes accumulation of lung macrophages and marked tissue fibrosis. Our study thus shows that PGE2 signalling, predominantly via EP4, plays an important role during the second wave of immune activity following resolution of inflammation. This secondary immune activation drives local tissue-resident T cell development while limiting tissue injury.


Assuntos
Dinoprostona , Modelos Animais de Doenças , Pulmão , Macrófagos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica , Receptores de Prostaglandina E Subtipo EP4 , Streptococcus pneumoniae , Animais , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/patologia , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/metabolismo , Camundongos , Dinoprostona/metabolismo , Streptococcus pneumoniae/imunologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Receptores de Prostaglandina E Subtipo EP4/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Pulmão/imunologia , Pulmão/patologia , Pulmão/microbiologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Cadeias alfa de Integrinas/metabolismo , Cadeias alfa de Integrinas/genética , Feminino , Antígenos CD/metabolismo , Antígenos CD/genética , Linfócitos T/imunologia
5.
Biochem Biophys Res Commun ; 715: 150007, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38678783

RESUMO

Smad4, a critical mediator of TGF-ß signaling, plays a pivotal role in regulating various cellular functions, including immune responses. In this study, we investigated the impact of Smad4 knockout specifically in macrophages on anti-tumor immunity, focusing on lung metastasis of B16 melanoma cells. Using a mouse model with Smad4 knockout in macrophages established via Lyz2-cre mice and Smad4 flox/flox mice, we demonstrated a significant inhibition of B16 metastasis in the lungs. Interestingly, the inhibition of tumor growth was found to be independent of adaptive immunity, as no significant changes were observed in the numbers or activities of T cells, B cells, or NK cells. Instead, Smad4 knockout led to the emergence of an MCHIIlow CD206high subset of lung interstitial macrophages, characterized by enhanced phagocytosis function. Our findings highlight the crucial role of Smad4 in modulating the innate immune response against tumors and provide insights into potential therapeutic strategies targeting lung interstitial macrophages to enhance anti-tumor immunity.


Assuntos
Neoplasias Pulmonares , Melanoma Experimental , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Proteína Smad4 , Animais , Proteína Smad4/deficiência , Proteína Smad4/genética , Proteína Smad4/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/genética , Melanoma Experimental/patologia , Melanoma Experimental/imunologia , Camundongos , Macrófagos/imunologia , Macrófagos/metabolismo , Pulmão/patologia , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/patologia , Linhagem Celular Tumoral
6.
Infect Immun ; 92(5): e0006024, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38619302

RESUMO

Melioidosis is an emerging tropical infection caused by inhalation, inoculation, or ingestion of the flagellated, facultatively intracellular pathogen Burkholderia pseudomallei. The melioidosis case fatality rate is often high, and pneumonia, the most common presentation, doubles the risk of death. The alveolar macrophage is a sentinel pulmonary host defense cell, but the human alveolar macrophage in B. pseudomallei infection has never been studied. The objective of this study was to investigate the host-pathogen interaction of B. pseudomallei infection with the human alveolar macrophage and to determine the role of flagellin in modulating inflammasome-mediated pathways. We found that B. pseudomallei infects primary human alveolar macrophages but is gradually restricted in the setting of concurrent cell death. Electron microscopy revealed cytosolic bacteria undergoing division, indicating that B. pseudomallei likely escapes the alveolar macrophage phagosome and may replicate in the cytosol, where it triggers immune responses. In paired human blood monocytes, uptake and intracellular restriction of B. pseudomallei are similar to those observed in alveolar macrophages, but cell death is reduced. The alveolar macrophage cytokine response to B. pseudomallei is characterized by marked interleukin (IL)-18 secretion compared to monocytes. Both cytotoxicity and IL-18 secretion in alveolar macrophages are partially flagellin dependent. However, the proportion of IL-18 release that is driven by flagellin is greater in alveolar macrophages than in monocytes. These findings suggest differential flagellin-mediated inflammasome pathway activation in the human alveolar macrophage response to B. pseudomallei infection and expand our understanding of intracellular pathogen recognition by this unique innate immune lung cell.


Assuntos
Burkholderia pseudomallei , Flagelina , Interações Hospedeiro-Patógeno , Inflamassomos , Macrófagos Alveolares , Humanos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Inflamassomos/imunologia , Inflamassomos/metabolismo , Burkholderia pseudomallei/imunologia , Flagelina/imunologia , Flagelina/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Melioidose/imunologia , Melioidose/microbiologia , Células Cultivadas
7.
Int Immunopharmacol ; 132: 111991, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38581996

RESUMO

OBJECTIVES: Acute lung injury (ALI) is a highly inflammatory condition with the involvement of M1 alveolar macrophages (AMs) polarization, eventually leading to the development of non-cardiogenic edema in alveolar and interstitial regions, accompanied by persistent hypoxemia. Given the significant mortality rate associated with ALI, it is imperative to investigate the underlying mechanisms of this condition so as to identify potential therapeutic targets. The therapeutic effects of the inhibition of bromodomain containing protein 4 (BRD4), an epigenetic reader, has been proven with high efficacy in ameliorating various inflammatory diseases through mediating immune cell activation. However, little is known about the therapeutic potential of BRD4 degradation in acute lung injury. METHODS: This study aimed to assess the protective efficacy of ARV-825, a novel BRD4-targeted proteolysis targeting chimera (PROTAC), against ALI through histopathological examination in lung tissues and biochemical analysis in bronchoalveolar lavage fluid (BALF). Additionally, the underlying mechanism by which BRD4 regulated M1 AMs was elucidated by using CUT & Tag assay. RESULTS: In this study, we found the upregulation of BRD4 in a lipopolysaccharide (LPS)-induced ALI model. Furthermore, we observed that intraperitoneal administration of ARV-825, significantly alleviated LPS-induced pulmonary pathological changes and inflammatory responses. These effects were accompanied by the suppression of M1 AMs. In addition, our findings revealed that the administration of ARV-825 effectively suppressed M1 AMs by inhibiting the expression of IRF7, a crucial transcriptional factor involved in M1 macrophages. CONCLUSION: Our study suggested that targeting BRD4 using ARV-825 is a potential therapeutic approach for ALI.


Assuntos
Lesão Pulmonar Aguda , Proteínas que Contêm Bromodomínio , Lipopolissacarídeos , Macrófagos Alveolares , Fatores de Transcrição , Animais , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/imunologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos , Masculino , Camundongos Endogâmicos C57BL , Humanos , Proteólise/efeitos dos fármacos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Pulmão/patologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Modelos Animais de Doenças , Ativação de Macrófagos/efeitos dos fármacos
8.
Microbiol Spectr ; 12(5): e0390523, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38501823

RESUMO

Pseudomonas aeruginosa (P. aeruginosa) is a Gram-negative facultative anaerobe that has become an important cause of severe infections in humans, particularly in patients with cystic fibrosis. The development of efficacious methods or mendicants against P. aeruginosa is still needed. We previously reported that regenerating islet-derived family member 4 (Reg4) has bactericidal activity against Salmonella Typhimurium, a Gram-negative flagellated bacterium. We herein explore whether Reg4 has bactericidal activity against P. aeruginosa. In the P. aeruginosa PAO1-chronic infection model, Reg4 significantly inhibits the colonization of PAO1 in the lung and subsequently ameliorates pulmonary inflammation and fibrosis. Reg4 recombinant protein suppresses the growth motility and biofilm formation capability of PAO1 in vitro. Mechanistically, Reg4 not only exerts bactericidal action via direct binding to the P. aeruginosa cell wall but also enhances the phagocytosis of alveolar macrophages in the host. Taken together, our study demonstrates that Reg4 may provide protection against P. aeruginosa-induced pulmonary inflammation and fibrosis via its antibacterial activity.IMPORTANCEChronic lung infection with Pseudomonas aeruginosa is a leading cause of morbidity and mortality in patients with cystic fibrosis. Due to the antibiotic resistance of Pseudomonas aeruginosa, antimicrobial peptides appear to be a potential alternative to combat its infection. In this study, we report an antimicrobial peptide, regenerating islet-derived 4 (Reg4), that showed killing activity against clinical strains of Pseudomonas aeruginosa PAO1 and ameliorated PAO1-induced pulmonary inflammation and fibrosis. Experimental data also showed Reg4 directly bound to the bacterial cell membrane and enhanced the phagocytosis of host alveolar macrophages. Our presented study will be a helpful resource in searching for novel antimicrobial peptides that could have the potential to replace conventional antibiotics.


Assuntos
Antibacterianos , Proteínas Associadas a Pancreatite , Infecções por Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/efeitos dos fármacos , Animais , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/tratamento farmacológico , Camundongos , Proteínas Associadas a Pancreatite/metabolismo , Proteínas Associadas a Pancreatite/genética , Antibacterianos/farmacologia , Humanos , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/imunologia , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Camundongos Endogâmicos C57BL , Pneumonia/microbiologia , Peptídeos Antimicrobianos/farmacologia , Fagocitose/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/patologia , Fibrose Cística/microbiologia , Fibrose Cística/complicações , Fibrose Pulmonar/microbiologia , Modelos Animais de Doenças
9.
Am J Physiol Lung Cell Mol Physiol ; 326(6): L672-L686, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38530936

RESUMO

Alveolar macrophages (AMs) in patients with chronic obstructive pulmonary disease (COPD) orchestrate persistent inflammation in the airway. However, subpopulations of AMs participating in chronic inflammation have been poorly characterized. We previously reported that Siglec-1 expression on AMs, which is important for bacteria engulfment, was decreased in COPD. Here, we show that Siglec-1-negative AMs isolated from COPD lung tissues exhibit a proinflammatory phenotype and are associated with poor clinical outcomes in patients with COPD. Using flow cytometry, we segregated three subsets of AMs based on the expression of Siglec-1 and their side scattergram (SSC) and forward scattergram (FSC) properties: Siglec-1+SSChiFSChi, Siglec-1-SSChiFSChi, and Siglec-1-SSCloFSClo subsets. The Siglec-1-SSCloFSClo subset number was increased in COPD. RNA sequencing revealed upregulation of multiple proinflammatory signaling pathways and emphysema-associated matrix metalloproteases in the Siglec-1-SSCloFSClo subset. Gene set enrichment analysis indicated that the Siglec-1-SSCloFSClo subset adopted intermediate phenotypes between monocytes and mature alveolar macrophages. Functionally, these cells produced TNF-α, IL-6, and IL-8 at baseline, and these cytokines were significantly increased in response to viral RNA. The increase in Siglec-1-negative AMs in induced sputum is associated with future exacerbation risk and lung function decline in patients with COPD. Collectively, the novel Siglec-1-SSCloFSClo subset of AMs displays proinflammatory properties, and their emergence in COPD airways may be associated with poor clinical outcomes.NEW & NOTEWORTHY Alveolar macrophages (AMs) in patients with chronic obstructive pulmonary disease (COPD) orchestrate persistent inflammation in the airway. We find that Siglec-1-negative alveolar macrophages have a wide range of proinflammatory landscapes and a protease-expressing phenotype. Moreover, this subset is associated with the pathogenesis of COPD and responds to viral stimuli.


Assuntos
Macrófagos Alveolares , Fenótipo , Doença Pulmonar Obstrutiva Crônica , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Macrófagos Alveolares/imunologia , Humanos , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Masculino , Feminino , Idoso , Pessoa de Meia-Idade , Inflamação/metabolismo , Inflamação/patologia , Citocinas/metabolismo
10.
J Biol Chem ; 300(4): 107199, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38508309

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV), a highly infectious virus, causes severe losses in the swine industry by regulating the inflammatory response, inducing tissue damage, suppressing the innate immune response, and promoting persistent infection in hosts. Interleukin-13 (IL-13) is a cytokine that plays a critical role in regulating immune responses and inflammation, particularly in immune-related disorders, certain types of cancer, and numerous bacterial and viral infections; however, the underlying mechanisms of IL-13 regulation during PRRSV infection are not well understood. In this study, we demonstrated that PRRSV infection elevates IL-13 levels in porcine alveolar macrophages. PRRSV enhances m6A-methylated RNA levels while reducing the expression of fat mass and obesity associated protein (FTO, an m6A demethylase), thereby augmenting IL-13 production. PRRSV nonstructural protein 9 (nsp9) was a key factor for this modulation. Furthermore, we found that the residues Asp567, Tyr586, Leu593, and Asp595 were essential for nsp9 to induce IL-13 production via attenuation of FTO expression. These insights delineate PRRSV nsp9's role in FTO-mediated IL-13 release, advancing our understanding of PRRSV's impact on host immune and inflammatory responses.


Assuntos
Interleucina-13 , Macrófagos Alveolares , Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Proteínas não Estruturais Virais , Animais , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Suínos , Interleucina-13/metabolismo , Interleucina-13/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virologia , Macrófagos Alveolares/imunologia , Síndrome Respiratória e Reprodutiva Suína/metabolismo , Síndrome Respiratória e Reprodutiva Suína/virologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Regulação para Cima
11.
Science ; 383(6685): eadi3808, 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38386728

RESUMO

Cancer risk is influenced by inherited mutations, DNA replication errors, and environmental factors. However, the influence of genetic variation in immunosurveillance on cancer risk is not well understood. Leveraging population-level data from the UK Biobank and FinnGen, we show that heterozygosity at the human leukocyte antigen (HLA)-II loci is associated with reduced lung cancer risk in smokers. Fine-mapping implicated amino acid heterozygosity in the HLA-II peptide binding groove in reduced lung cancer risk, and single-cell analyses showed that smoking drives enrichment of proinflammatory lung macrophages and HLA-II+ epithelial cells. In lung cancer, widespread loss of HLA-II heterozygosity (LOH) favored loss of alleles with larger neopeptide repertoires. Thus, our findings nominate genetic variation in immunosurveillance as a critical risk factor for lung cancer.


Assuntos
Predisposição Genética para Doença , Antígenos de Histocompatibilidade Classe II , Vigilância Imunológica , Perda de Heterozigosidade , Neoplasias Pulmonares , Humanos , Antígenos de Histocompatibilidade Classe II/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Macrófagos Alveolares/imunologia , Fatores de Risco , Fumar/imunologia , Vigilância Imunológica/genética , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Mapeamento Cromossômico , Polimorfismo de Nucleotídeo Único
12.
J Biol Chem ; 300(1): 105518, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38042489

RESUMO

Bacillus Calmette-Guérin (BCG) vaccination induces a type of immune memory known as "trained immunity", characterized by the immunometabolic and epigenetic changes in innate immune cells. However, the molecular mechanism underlying the strategies for inducing and/or boosting trained immunity in alveolar macrophages remains unknown. Here, we found that mucosal vaccination with the recombinant strain rBCGPPE27 significantly augmented the trained immune response in mice, facilitating a superior protective response against Mycobacterium tuberculosis and non-related bacterial reinfection in mice when compared to BCG. Mucosal immunization with rBCGPPE27 enhanced innate cytokine production by alveolar macrophages associated with promoted glycolytic metabolism, typical of trained immunity. Deficiency of the mammalian target of rapamycin complex 2 and hexokinase 1 abolished the immunometabolic and epigenetic rewiring in mouse alveolar macrophages after mucosal rBCGPPE27 vaccination. Most noteworthy, utilizing rBCGPPE27's higher-up trained effects: The single mucosal immunization with rBCGPPE27-adjuvanted coronavirus disease (CoV-2) vaccine raised the rapid development of virus-specific immunoglobulin G antibodies, boosted pseudovirus neutralizing antibodies, and augmented T helper type 1-biased cytokine release by vaccine-specific T cells, compared to BCG/CoV-2 vaccine. These findings revealed that mucosal recombinant BCG vaccine induces lung-resident memory macrophages and enhances trained immunity via reprogramming mTORC2- and HK-1-mediated aerobic glycolysis, providing new vaccine strategies for improving tuberculosis (TB) or coronavirus variant vaccinations, and targeting innate immunity via mucosal surfaces.


Assuntos
Vacina BCG , Hexoquinase , Memória Imunológica , Pulmão , Macrófagos Alveolares , Alvo Mecanístico do Complexo 2 de Rapamicina , Mycobacterium tuberculosis , Imunidade Treinada , Animais , Camundongos , Vacina BCG/imunologia , Citocinas/metabolismo , Pulmão/imunologia , Macrófagos Alveolares/imunologia , Mycobacterium tuberculosis/imunologia , Vacinas Sintéticas/imunologia , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Hexoquinase/metabolismo
13.
PLoS Pathog ; 19(7): e1011556, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37498977

RESUMO

Although alveolar macrophages (AMs) play important roles in preventing and eliminating pulmonary infections, little is known about their regulation in healthy animals. Since exposure to LPS often renders cells hyporesponsive to subsequent LPS exposures ("tolerant"), we tested the hypothesis that LPS produced in the intestine reaches the lungs and stimulates AMs, rendering them tolerant. We found that resting AMs were more likely to be tolerant in mice lacking acyloxyacyl hydrolase (AOAH), the host lipase that degrades and inactivates LPS; isolated Aoah-/- AMs were less responsive to LPS stimulation and less phagocytic than were Aoah+/+ AMs. Upon innate stimulation in the airways, Aoah-/- mice had reduced epithelium- and macrophage-derived chemokine/cytokine production. Aoah-/- mice also developed greater and more prolonged loss of body weight and higher bacterial burdens after pulmonary challenge with Pseudomonas aeruginosa than did wildtype mice. We also found that bloodborne or intrarectally-administered LPS desensitized ("tolerized") AMs while antimicrobial drug treatment that reduced intestinal commensal Gram-negative bacterial abundance largely restored the innate responsiveness of Aoah-/- AMs. Confirming the role of LPS stimulation, the absence of TLR4 prevented Aoah-/- AM tolerance. We conclude that commensal LPSs may stimulate and desensitize (tolerize) alveolar macrophages in a TLR4-dependent manner and compromise pulmonary immunity. By inactivating LPS in the intestine, AOAH promotes antibacterial host defenses in the lung.


Assuntos
Hidrolases de Éster Carboxílico , Macrófagos Alveolares , Animais , Camundongos , Lipopolissacarídeos/toxicidade , Pulmão , Macrófagos Alveolares/imunologia , Receptor 4 Toll-Like , Hidrolases de Éster Carboxílico/metabolismo
15.
Cancer Res ; 83(12): 2016-2033, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37078793

RESUMO

Cellular components of the tumor microenvironment, including myeloid cells, play important roles in the progression of lung adenocarcinoma (LUAD) and its response to therapy. Here, we characterize the function of the ubiquitin ligases Siah1a/2 in regulating the differentiation and activity of alveolar macrophages (AM) and assess the implication of Siah1a/2 control of AMs for carcinogen-induced LUAD. Macrophage-specific genetic ablation of Siah1a/2 promoted accumulation of AMs with an immature phenotype and increased expression of protumorigenic and pro-inflammatory Stat3 and ß-catenin gene signatures. Administration of urethane to wild-type mice promoted enrichment of immature-like AMs and lung tumor development, which was enhanced by macrophage-specific Siah1a/2 ablation. The profibrotic gene signature seen in Siah1a/2-ablated immature-like macrophages was associated with increased tumor infiltration of CD14+ myeloid cells and poorer survival of patients with LUAD. Single-cell RNA-seq confirmed the presence of a cluster of immature-like AMs expressing a profibrotic signature in lungs of patients with LUAD, a signature enhanced in smokers. These findings identify Siah1a/2 in AMs as gatekeepers of lung cancer development. SIGNIFICANCE: The ubiquitin ligases Siah1a/2 control proinflammatory signaling, differentiation, and profibrotic phenotypes of alveolar macrophages to suppress lung carcinogenesis.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Macrófagos Alveolares , Ubiquitina-Proteína Ligases , Animais , Camundongos , Macrófagos Alveolares/enzimologia , Macrófagos Alveolares/imunologia , Adenocarcinoma de Pulmão/induzido quimicamente , Neoplasias Pulmonares/induzido quimicamente , Microambiente Tumoral , Ubiquitina-Proteína Ligases/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout
16.
Front Immunol ; 13: 877383, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35844541

RESUMO

The mold Alternaria alternata is one of the main sources of asthma exacerbation, being its major allergen, Alt a 1, indispensable for its development. The main objective of this work was to answer two main questions: 1) can Alt a 1 by itself (without any other context) induce an asthmatic profile in vivo?; and 2) Which molecular mechanisms take place during this phenomenon? To answer both questions, we have developed a mouse model of allergic asthma using only Alt a 1 for mice sensitization. We also made use of in-vitro cellular models and computational studies to support some aspects of our hypothesis. Our results showed that Alt a 1 can induce an asthmatic phenotype, promoting tissue remodeling and infiltration of CD45+ cells, especially eosinophils and macrophages (Siglec F+ and F4/80+). Also, we have found that Alt a 1 sensitization is mediated by the TLR4-macrophage axis.


Assuntos
Asma , Proteínas Fúngicas , Macrófagos Alveolares , Receptor 4 Toll-Like , Alérgenos , Animais , Asma/imunologia , Eosinófilos/imunologia , Proteínas Fúngicas/imunologia , Macrófagos Alveolares/imunologia , Camundongos , Receptor 4 Toll-Like/imunologia
17.
Vet Pathol ; 59(4): 627-638, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35499307

RESUMO

Emerging and re-emerging human coronaviruses (hCoVs) cause severe respiratory illness in humans, but the basis for lethal pneumonia in these diseases is not well understood. Alveolar macrophages (AMs) are key orchestrators of host antiviral defense and tissue tolerance during a variety of respiratory infections, and AM dysfunction is associated with severe COVID-19. In this study, using a mouse model of Middle East respiratory syndrome coronavirus (MERS-CoV) infection, we examined the role of AMs in MERS pathogenesis. Our results show that depletion of AMs using clodronate (CL) liposomes significantly increased morbidity and mortality in human dipeptidyl peptidase 4 knock-in (hDPP4-KI) mice. Detailed examination of control and AM-depleted lungs at different days postinfection revealed increased neutrophil activity but a significantly reduced MERS-CoV-specific CD4 T-cell response in AM-deficient lungs during later stages of infection. Furthermore, enhanced MERS severity in AM-depleted mice correlated with lung inflammation and lesions. Collectively, these data demonstrate that AMs are critical for the development of an optimal virus-specific T-cell response and controlling excessive inflammation during MERS-CoV infection.


Assuntos
Infecções por Coronavirus , Macrófagos Alveolares , Coronavírus da Síndrome Respiratória do Oriente Médio , Pneumonia , Animais , Ácido Clodrônico , Infecções por Coronavirus/imunologia , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Transgênicos , Pneumonia/imunologia , Pneumonia/virologia
18.
Proc Natl Acad Sci U S A ; 119(15): e2121098119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35377803

RESUMO

The pathogenesis of lung fibrosis involves hyperactivation of innate and adaptive immune pathways that release inflammatory cytokines and growth factors such as tumor growth factor (TGF)ß1 and induce aberrant extracellular matrix protein production. During the genesis of pulmonary fibrosis, resident alveolar macrophages are replaced by a population of newly arrived monocyte-derived interstitial macrophages that subsequently transition into alveolar macrophages (Mo-AMs). These transitioning cells initiate fibrosis by releasing profibrotic cytokines and remodeling the matrix. Here, we describe a strategy for leveraging the up-regulation of the mannose receptor CD206 in interstitial macrophages and Mo-AM to treat lung fibrosis. We engineered mannosylated albumin nanoparticles, which were found to be internalized by fibrogenic CD206+ monocyte derived macrophages (Mo-Macs). Mannosylated albumin nanoparticles incorporating TGFß1 small-interfering RNA (siRNA) targeted the profibrotic subpopulation of CD206+ macrophages and prevented lung fibrosis. The findings point to the potential utility of mannosylated albumin nanoparticles in delivering TGFß-siRNA into CD206+ profibrotic macrophages as an antilung fibrosis strategy.


Assuntos
Linfotoxina-alfa , Macrófagos Alveolares , Nanopartículas , Fibrose Pulmonar , RNA Interferente Pequeno , Animais , Bleomicina/farmacologia , Modelos Animais de Doenças , Linfotoxina-alfa/genética , Macrófagos Alveolares/imunologia , Receptor de Manose , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/terapia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética
19.
J Leukoc Biol ; 112(3): 499-512, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35435271

RESUMO

Clearance of airway intruders by immune cells is required to resolve infectious pneumonia. However, the molecular mechanisms underlying this process remain elusive. Here, we demonstrated that alveolar macrophage (AM)-derived neuropilin 2 (NRP2) plays an essential role in controlling severe pneumonia by enhancing microbial clearance. Mice with conditional deletion of the NRP2 gene in AM had persistent bacteria, uncontrolled neutrophil influx, and decreased survival during Escherichia coli-induced pneumonia. In vitro assays demonstrated that NRP2 could bind to CD11b+ Ly6Glo/+ neutrophils and promote their capacities in phagocytosis and killing of bacteria, which is partially contributed to the increased expression of TLR4 and TNF-a. These findings collectively revealed that AM-derived NRP2 protects the lungs from unwanted injury by promoting the clearance of invading pathogens. This study might provide a promising diagnostic biomarker and therapeutic target for severe pneumonia.


Assuntos
Infecções por Escherichia coli , Macrófagos Alveolares , Neuropilina-2 , Pneumonia Bacteriana , Animais , Infecções por Escherichia coli/imunologia , Pulmão , Lesão Pulmonar , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neuropilina-2/metabolismo , Neutrófilos , Fagocitose , Pneumonia Bacteriana/imunologia
20.
Commun Biol ; 5(1): 242, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35304580

RESUMO

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), has incited a global health crisis. Currently, there are limited therapeutic options for the prevention and treatment of SARS-CoV-2 infections. We evaluated the antiviral activity of sulforaphane (SFN), the principal biologically active phytochemical derived from glucoraphanin, the naturally occurring precursor present in high concentrations in cruciferous vegetables. SFN inhibited in vitro replication of six strains of SARS-CoV-2, including Delta and Omicron, as well as that of the seasonal coronavirus HCoV-OC43. Further, SFN and remdesivir interacted synergistically to inhibit coronavirus infection in vitro. Prophylactic administration of SFN to K18-hACE2 mice prior to intranasal SARS-CoV-2 infection significantly decreased the viral load in the lungs and upper respiratory tract and reduced lung injury and pulmonary pathology compared to untreated infected mice. SFN treatment diminished immune cell activation in the lungs, including significantly lower recruitment of myeloid cells and a reduction in T cell activation and cytokine production. Our results suggest that SFN should be explored as a potential agent for the prevention or treatment of coronavirus infections.


Assuntos
Antivirais/uso terapêutico , Resfriado Comum/tratamento farmacológico , Infecções por Coronavirus/tratamento farmacológico , Coronavirus Humano OC43 , Isotiocianatos/uso terapêutico , SARS-CoV-2 , Sulfóxidos/uso terapêutico , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/uso terapêutico , Alanina/análogos & derivados , Alanina/uso terapêutico , Animais , Células CACO-2 , Chlorocebus aethiops , Resfriado Comum/virologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Citocinas/imunologia , Sinergismo Farmacológico , Humanos , Pulmão/imunologia , Pulmão/virologia , Macrófagos Alveolares/imunologia , Masculino , Camundongos Transgênicos , Baço/imunologia , Linfócitos T/imunologia , Células Vero , Carga Viral , Tratamento Farmacológico da COVID-19
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA