Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 844
Filtrar
1.
Nat Microbiol ; 9(4): 949-963, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38528148

RESUMEN

A polymorphism causing deficiencies in Toll-interacting protein (TOLLIP), an inhibitory adaptor protein affecting endosomal trafficking, is associated with increased tuberculosis (TB) risk. It is, however, unclear how TOLLIP affects TB pathogenesis. Here we show that TB severity is increased in Tollip-/- mice, characterized by macrophage- and T cell-driven inflammation, foam cell formation and lipid accumulation. Tollip-/- alveolar macrophages (AM) specifically accumulated lipid and underwent necrosis. Transcriptional and protein analyses of Mycobacterium tuberculosis (Mtb)-infected, Tollip-/- AM revealed increased EIF2 signalling and downstream upregulation of the integrated stress response (ISR). These phenotypes were linked, as incubation of the Mtb lipid mycolic acid with Mtb-infected Tollip-/- AM activated the ISR and increased Mtb replication. Correspondingly, the ISR inhibitor, ISRIB, reduced Mtb numbers in AM and improved Mtb control, overcoming the inflammatory phenotype. In conclusion, targeting the ISR offers a promising target for host-directed anti-TB therapy towards improved Mtb control and reduced immunopathology.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Ratones , Macrófagos Alveolares/microbiología , Tuberculosis/microbiología , Mycobacterium tuberculosis/fisiología , Macrófagos/microbiología , Lípidos , Péptidos y Proteínas de Señalización Intracelular/metabolismo
2.
Front Cell Infect Microbiol ; 13: 1266884, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38029268

RESUMEN

Tuberculosis (TB), attributed to the Mycobacterium tuberculosis complex, is one of the most serious zoonotic diseases worldwide. Nevertheless, the host mechanisms preferentially leveraged by Mycobacterium remain unclear. After infection, both Mycobacterium tuberculosis (MTB) and Mycobacterium bovis (MB) bacteria exhibit intimate interactions with host alveolar macrophages; however, the specific mechanisms underlying these macrophage responses remain ambiguous. In our study, we performed a comparative proteomic analysis of bovine alveolar macrophages (BAMs) infected with MTB or MB to elucidate the differential responses of BAMs to each pathogen at the protein level. Our findings revealed heightened TB infection susceptibility of BAMs that had been previously infected with MTB or MB. Moreover, we observed that both types of mycobacteria triggered significant changes in BAM energy metabolism. A variety of proteins and signalling pathways associated with autophagy and inflammation-related progression were highly activated in BAMs following MB infection. Additionally, proteins linked to energy metabolism were highly expressed in BAMs following MTB infection. In summary, we propose that BAMs may resist MTB and MB infections via different mechanisms. Our findings provide critical insights into TB pathogenesis, unveiling potential biomarkers to facilitate more effective TB treatment strategies. Additionally, our data lend support to the hypothesis that MTB may be transmitted via cross-species infection.


Asunto(s)
Mycobacterium bovis , Mycobacterium tuberculosis , Tuberculosis , Animales , Bovinos , Mycobacterium tuberculosis/fisiología , Macrófagos Alveolares/microbiología , Proteoma , Proteómica , Tuberculosis/veterinaria
3.
Molecules ; 28(17)2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37687052

RESUMEN

Secretory phospholipase B1 (PLB1) and biofilms act as microbial virulence factors and play an important role in pulmonary cryptococcosis. This study aims to formulate the ethanolic extract of propolis-loaded niosomes (Nio-EEP) and evaluate the biological activities occurring during PLB1 production and biofilm formation of Cryptococcus neoformans. Some physicochemical characterizations of niosomes include a mean diameter of 270 nm in a spherical shape, a zeta-potential of -10.54 ± 1.37 mV, and 88.13 ± 0.01% entrapment efficiency. Nio-EEP can release EEP in a sustained manner and retains consistent physicochemical properties for a month. Nio-EEP has the capability to permeate the cellular membranes of C. neoformans, causing a significant decrease in the mRNA expression level of PLB1. Interestingly, biofilm formation, biofilm thickness, and the expression level of biofilm-related genes (UGD1 and UXS1) were also significantly reduced. Pre-treating with Nio-EEP prior to yeast infection reduced the intracellular replication of C. neoformans in alveolar macrophages by 47%. In conclusion, Nio-EEP mediates as an anti-virulence agent to inhibit PLB1 and biofilm production for preventing fungal colonization on lung epithelial cells and also decreases the intracellular replication of phagocytosed cryptococci. This nano-based EEP delivery might be a potential therapeutic strategy in the prophylaxis and treatment of pulmonary cryptococcosis in the future.


Asunto(s)
Antifúngicos , Biopelículas , Cryptococcus neoformans , Proteínas Fúngicas , Lisofosfolipasa , Macrófagos Alveolares , Própolis , Humanos , Biopelículas/efectos de los fármacos , Línea Celular Tumoral , Criptococosis/prevención & control , Criptococosis/terapia , Cryptococcus neoformans/efectos de los fármacos , Cryptococcus neoformans/enzimología , Cryptococcus neoformans/patogenicidad , Etanol/química , Proteínas Fúngicas/antagonistas & inhibidores , Liposomas , Enfermedades Pulmonares Fúngicas/prevención & control , Enfermedades Pulmonares Fúngicas/terapia , Lisofosfolipasa/antagonistas & inhibidores , Macrófagos Alveolares/microbiología , Própolis/química , Própolis/farmacología , Virulencia/efectos de los fármacos , Factores de Virulencia/antagonistas & inhibidores , Antifúngicos/química , Antifúngicos/farmacología
4.
Front Cell Infect Microbiol ; 13: 1062963, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36936766

RESUMEN

Mycobacterium tuberculosis (Mtb), the causative agent of Tuberculosis (TB), remains a pathogen of great interest on a global scale. This airborne pathogen affects the lungs, where it interacts with macrophages. Acidic pH, oxidative and nitrosative stressors, and food restrictions make the macrophage's internal milieu unfriendly to foreign bodies. Mtb subverts the host immune system and causes infection due to its genetic arsenal and secreted effector proteins. In vivo and in vitro research have examined Mtb-host macrophage interaction. This interaction is a crucial stage in Mtb infection because lung macrophages are the first immune cells Mtb encounters in the host. This review summarizes Mtb effectors that interact with macrophages. It also examines how macrophages control and eliminate Mtb and how Mtb manipulates macrophage defense mechanisms for its own survival. Understanding these mechanisms is crucial for TB prevention, diagnosis, and treatment.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Mycobacterium tuberculosis/genética , Macrófagos/microbiología , Tuberculosis/microbiología , Macrófagos Alveolares/microbiología , Pulmón/microbiología , Interacciones Huésped-Patógeno
5.
Infect Immun ; 91(4): e0049022, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-36916933

RESUMEN

Pneumocystis is a respiratory fungal pathogen that is among the most frequent causes of life-threatening pneumonia (PcP) in immunocompromised hosts. Alveolar macrophages play an important role in host defense against Pneumocystis, and several studies have suggested that M2 polarized macrophages have anti-Pneumocystis effector activity. Our prior work found that the immunomodulatory drug sulfasalazine (SSZ) provides a dual benefit during PcP-related immune reconstitution inflammatory syndrome (IRIS) by concurrently suppressing immunopathogenesis while also accelerating macrophage-mediated fungal clearance. The benefits of SSZ were associated with heightened Th2 cytokine production and M2 macrophage polarization. Therefore, to determine whether SSZ improves the outcome of PcP through a mechanism that requires Th2-dependent M2 polarization, RAG2-/- mice lacking interleukin 4 receptor alpha chain (IL-4Rα) on macrophage lineage cells were generated. As expected, SSZ treatment dramatically reduced the severity of PcP-related immunopathogenesis and accelerated fungal clearance in immune-reconstituted RAG2-/- mice. Similarly, SSZ treatment was also highly effective in immune-reconstituted RAG2/IL-4Rα-/- and RAG2/gamma interferon receptor (IFN-γR)-/- mice, demonstrating that neither IL-4Rα-dependent M2 nor IFN-γR-dependent M1 macrophage polarization programs were required for the beneficial effects of SSZ. Despite the fact that macrophages from RAG2/IL-4Rα-/- mice could not respond to the Th2 cytokines IL-4 and IL-13, M2-biased alveolar macrophages were identified in the lungs following SSZ treatment. These data demonstrate that not only does SSZ enhance phagocytosis and fungal clearance in the absence of macrophage IL-4Rα signaling, but also that SSZ promotes M2 macrophage polarization in an IL-4Rα-independent manner. These findings could have implications for the treatment of PcP and other diseases in which M2 polarization is beneficial.


Asunto(s)
Pneumocystis , Neumonía por Pneumocystis , Ratones , Animales , Sulfasalazina/farmacología , Neumonía por Pneumocystis/tratamiento farmacológico , Antifúngicos/farmacología , Macrófagos , Macrófagos Alveolares/microbiología
6.
Virulence ; 14(1): 2171636, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-36694280

RESUMEN

Haemophilus parasuis (H. parasuis) causes exudative inflammation, implying endothelial dysfunction during pathogen infection. However, so far, the molecular mechanism of endothelial dysfunction caused by H. parasuis has not been clarified. By using the transwell-based cell co-culture system, we demonstrate that knocking out resistin in porcine alveolar macrophages (PAMs) dramatically attenuated endothelial monolayer damage caused by H. parasuis. The resistin secreted by PAMs inhibited the expression of the tight junction proteins claudin-5 and occludin rather than the adherens junction protein VE-cadherin in co-cultured porcine aortic endothelial cells (PAECs). Furthermore, we demonstrate that resistin regulated claudin-5 and occludin expression and monolayer PAEC permeability in an LKB1/AMPK/mTOR pathway-dependent manner. Additionally, we reveal that the outer membrane lipoprotein gene lppA in H. parasuis induced resistin expression in PAMs, as deleting lppA reduced resistin expression in H. parasuis-infected PAMs, causing a significant change in LKB1/AMPK/mTOR pathway activity in co-cultured PAECs, thereby restoring tight junction protein levels and endothelial monolayer permeability. Thus, we postulate that the H. parasuis lppA gene enhances resistin production in PAMs, disrupting tight junctions in PAECs and causing endothelial barrier dysfunction. These findings elucidate the pathogenic mechanism of exudative inflammation caused by H. parasuis for the first time and provide a more profound angle of acute exudative inflammation caused by bacteria.


Asunto(s)
Infecciones por Haemophilus , Haemophilus parasuis , Porcinos , Animales , Macrófagos Alveolares/microbiología , Haemophilus parasuis/genética , Células Endoteliales , Resistina/genética , Resistina/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Claudina-5/metabolismo , Ocludina/metabolismo , Infecciones por Haemophilus/veterinaria , Infecciones por Haemophilus/microbiología , Inflamación , Serina-Treonina Quinasas TOR/metabolismo
7.
J Immunol ; 210(4): 431-441, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36602769

RESUMEN

In Mycobacterium avium infections, macrophages play a critical role in the host defense response. Apoptosis inhibitor of macrophage (AIM), also known as CD5L, may represent a novel supportive therapy against various diseases, including metabolic syndrome and infectious diseases. The mechanisms of AIM include modulating lipid metabolism in macrophages and other host cells. We investigated the role of AIM in M. avium infections in vitro and in vivo. In a mouse model of M. avium pneumonia, foamy macrophages were induced 6 wk after infection. The bacteria localized in these macrophages. Flow cytometric analysis also confirmed that the percentage of CD11chighMHCclassIIhigh interstitial and alveolar macrophages, a cell surface marker defined as foamy macrophages, increased significantly after infection. AIM in alveolar lavage fluid and serum gradually increased after infection. Administration of recombinant AIM significantly increased the number of bacteria in the lungs of mice, accompanied by the induction of inflammatory cytokine and iNOS expression. In mouse bone marrow-derived macrophages, the mRNA expression of AIM after M. avium infection and the amount of AIM in the supernatant increased prior to the increase in intracellular bacteria. Infected cells treated with anti-AIM Abs had fewer bacteria and a higher percentage of apoptosis-positive cells than infected cells treated with isotype control Abs. Finally, AIM in the sera of patients with M. avium-pulmonary disease was measured and was significantly higher than in healthy volunteers. This suggests that AIM production is enhanced in M. avium-infected macrophages, increasing macrophage resistance to apoptosis and providing a possible site for bacterial growth.


Asunto(s)
Infección por Mycobacterium avium-intracellulare , Mycobacterium avium , Ratones , Animales , Macrófagos/fisiología , Infección por Mycobacterium avium-intracellulare/complicaciones , Infección por Mycobacterium avium-intracellulare/microbiología , Macrófagos Alveolares/microbiología , Apoptosis
8.
Int Immunopharmacol ; 104: 108407, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34924313

RESUMEN

Mycobacterium tuberculosis (M. tuberculosis (MTB) and M. bovis (MB) of the Mycobacterium tuberculosis complex (MTBC) are the causative agents of the notorious infectious disease tuberculosis (TB) in a range of mammals, including bovine and human. The lipid composition of MTB/MB performed imperative function as invading host macrophage. However, the detailed variations in lipid compositions of MTB and MB were unknown, while the responses relevant to lipid metabolisms in MTB/MB-infected host were also unclear. In the present study, a dual-Lipidomics were used to elucidate the differences in lipid composition of MTB and MB and responses in lipid metabolisms of primary bovine alveolar macrophages infected by MTB/MB. The Lipidomics showed significant differences in lipid composition, especially differences in levels of Glycerophospholipids, Sterol Lipids, Fatty Acyls and Polyketides between these two mycobacterium species. Meanwhile, both MTB and MB could invoke various responses of lipid metabolisms in host macrophages. An infection of MTB mainly induced the increases of Polyketides and Glycerophospholipids in macrophages, whereas an MB infection induced the increases of Glycerophospholipids and Sterol. Furthermore, TAG 13:0-18:5-18:5 of MTB and PC (16:1(9E)/0:0), PI(20:2(11Z,14Z)/22:6(4Z,7Z,10Z,13Z,16Z,19Z)), 4, 6-Decadiyn-1-ol isovalerate and LacCer (d18:1/24:1(15Z)) of MB were identified to cause variations in lipid metabolisms of macrophages, respectively. From these data, we proposed that the differential compositions of lipid compositions in MTB and MB could successfully colonize in macrophage by different mechanisms. MTB could promote the formation of foam cells of macrophage for its colonization and development, while MB mainly suppresses the macrophage autophagy to escape the immune responses of host.


Asunto(s)
Metabolismo de los Lípidos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiología , Mycobacterium bovis/metabolismo , Mycobacterium tuberculosis/metabolismo , Animales , Bovinos , Células Espumosas , Lipidómica , Lípidos/análisis , Masculino , Tuberculosis/metabolismo , Tuberculosis/veterinaria
9.
Front Immunol ; 12: 742370, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745115

RESUMEN

Tuberculosis is a deadly, contagious respiratory disease that is caused by the pathogenic bacterium Mycobacterium tuberculosis (Mtb). Mtb is adept at manipulating and evading host immunity by hijacking alveolar macrophages, the first line of defense against inhaled pathogens, by regulating the mode and timing of host cell death. It is established that Mtb infection actively blocks apoptosis and instead induces necrotic-like modes of cell death to promote disease progression. This survival strategy shields the bacteria from destruction by the immune system and antibiotics while allowing for the spread of bacteria at opportunistic times. As such, it is critical to understand how Mtb interacts with host macrophages to manipulate the mode of cell death. Herein, we demonstrate that Mtb infection triggers a time-dependent reduction in the expression of focal adhesion kinase (FAK) in human macrophages. Using pharmacological perturbations, we show that inhibition of FAK (FAKi) triggers an increase in a necrotic form of cell death during Mtb infection. In contrast, genetic overexpression of FAK (FAK+) completely blocked macrophage cell death during Mtb infection. Using specific inhibitors of necrotic cell death, we show that FAK-mediated cell death during Mtb infection occurs in a RIPK1-depedent, and to a lesser extent, RIPK3-MLKL-dependent mechanism. Consistent with these findings, FAKi results in uncontrolled replication of Mtb, whereas FAK+ reduces the intracellular survival of Mtb in macrophages. In addition, we demonstrate that enhanced control of intracellular Mtb replication by FAK+ macrophages is a result of increased production of antibacterial reactive oxygen species (ROS) as inhibitors of ROS production restored Mtb burden in FAK+ macrophages to same levels as in wild-type cells. Collectively, our data establishes FAK as an important host protective response during Mtb infection to block necrotic cell death and induce ROS production, which are required to restrict the survival of Mtb.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Interacciones Huésped-Patógeno/fisiología , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/patología , Tuberculosis Pulmonar/inmunología , Línea Celular , Humanos , Macrófagos Alveolares/enzimología , Mycobacterium tuberculosis/inmunología , Necrosis/inmunología , Especies Reactivas de Oxígeno/metabolismo
10.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34768852

RESUMEN

Given the rise of morbidity and mortality caused by Klebsiella pneumoniae (KP), the increasing number of strains resistant to antibiotics, and the emergence of hypervirulent Klebsiella pneumonia, treatment of KP infection becomes difficult; thus, novel drugs are necessary for treatment. Anthocyanins, or natural flavonoids, have an extensive effect against bacterial infection. However, few studies on anti-KP are identified. Here, we evaluated the therapeutic effect of purple sweet potato anthocyanins (PSPAs) on KP, containing 98.7% delphinidin 3-sambubioside. Results showed that KP-infected mice after PSPAs treatment manifested decreased mortality, weakened lung injury, dampened inflammatory responses, and reduced bacterial systemic dissemination in vivo. In Vitro, PSPAs significantly suppressed pyroptosis and restricted NLRP3 inflammasome activation in alveolar macrophages infected with KP. As for the mechanism, PSPAs promote mitophagy by recruiting Parkin to the mitochondria. PSPAs-conferred mitophagy increased mitochondrial membrane potential and decreased mitochondrial reactive oxygen species and mitochondrial DNA, resulting in impaired NLRP3 inflammasome activation. In addition, the promotion of mitophagy by PSPAs required the Nrf2 signaling pathway. Collectively, these findings suggest that PSPAs are a potential option for the treatment of KP infection.


Asunto(s)
Antocianinas/farmacología , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae/efectos de los fármacos , Mitofagia/efectos de los fármacos , Piroptosis/efectos de los fármacos , Animales , Antocianinas/análisis , Antocianinas/química , Línea Celular , ADN Mitocondrial/genética , Modelos Animales de Enfermedad , Femenino , Inflamación/tratamiento farmacológico , Ipomoea batatas/química , Klebsiella pneumoniae/metabolismo , Klebsiella pneumoniae/patogenicidad , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/prevención & control , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Extractos Vegetales/farmacología , Especies Reactivas de Oxígeno/metabolismo
11.
Front Immunol ; 12: 738070, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777348

RESUMEN

Mycobacterium abscessus (MAB) is one of the rapidly growing, multidrug-resistant non-tuberculous mycobacteria (NTM) causing various diseases including pulmonary disorder. Although it has been known that type I interferons (IFNs) contribute to host defense against bacterial infections, the role of type I IFNs against MAB infection is still unclear. In the present study, we show that rIFN-ß treatment reduced the intracellular growth of MAB in macrophages. Deficiency of IFN-α/ß receptor (IFNAR) led to the reduction of nitric oxide (NO) production in MAB-infected macrophages. Consistently, rIFN-ß treatment enhanced the expression of iNOS gene and protein, and NO production in response to MAB. We also found that NO is essential for the intracellular growth control of MAB within macrophages in an inhibitor assay using iNOS-deficient cells. In addition, pretreatment of rIFN-ß before MAB infection in mice increased production of NO in the lungs at day 1 after infection and promoted the bacterial clearance at day 5. However, when alveolar macrophages were depleted by treatment of clodronate liposome, rIFN-ß did not promote the bacterial clearance in the lungs. Moreover, we found that a cytosolic receptor nucleotide-binding oligomerization domain 2 (NOD2) is required for MAB-induced TANK binding kinase 1 (TBK1) phosphorylation and IFN-ß gene expression in macrophages. Finally, increase in the bacterial loads caused by reduction of NO levels was reversed by rIFN-ß treatment in the lungs of NOD2-deficient mice. Collectively, our findings suggest that type I IFNs act as an intermediator of NOD2-induced NO production in macrophages and thus contribute to host defense against MAB infection.


Asunto(s)
Interferón Tipo I/metabolismo , Pulmón/microbiología , Macrófagos Alveolares/microbiología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium abscessus/crecimiento & desarrollo , Óxido Nítrico/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Mycobacterium no Tuberculosas/inmunología , Infecciones por Mycobacterium no Tuberculosas/metabolismo , Mycobacterium abscessus/inmunología , Mycobacterium abscessus/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal
12.
Elife ; 102021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34544549

RESUMEN

Vaccination strategies for rapid protection against multidrug-resistant bacterial infection are very important, especially for hospitalized patients who have high risk of exposure to these bacteria. However, few such vaccination strategies exist due to a shortage of knowledge supporting their rapid effect. Here, we demonstrated that a single intranasal immunization of inactivated whole cell of Acinetobacter baumannii elicits rapid protection against broad A. baumannii-infected pneumonia via training of innate immune response in Rag1-/- mice. Immunization-trained alveolar macrophages (AMs) showed enhanced TNF-α production upon restimulation. Adoptive transfer of immunization-trained AMs into naive mice mediated rapid protection against infection. Elevated TLR4 expression on vaccination-trained AMs contributed to rapid protection. Moreover, immunization-induced rapid protection was also seen in Pseudomonas aeruginosa and Klebsiella pneumoniae pneumonia models, but not in Staphylococcus aureus and Streptococcus pneumoniae model. Our data reveal that a single intranasal immunization induces rapid and efficient protection against certain Gram-negative bacterial pneumonia via training AMs response, which highlights the importance and the possibility of harnessing trained immunity of AMs to design rapid-effecting vaccine.


Asunto(s)
Infecciones por Acinetobacter/prevención & control , Acinetobacter baumannii/inmunología , Vacunas Bacterianas/administración & dosificación , Infecciones por Klebsiella/prevención & control , Klebsiella pneumoniae/inmunología , Macrófagos Alveolares/efectos de los fármacos , Neumonía Bacteriana/prevención & control , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/inmunología , Infecciones por Acinetobacter/inmunología , Infecciones por Acinetobacter/microbiología , Administración Intranasal , Traslado Adoptivo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Proteínas de Homeodominio/genética , Inmunidad Innata/efectos de los fármacos , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/microbiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/trasplante , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Factores de Tiempo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Vacunación , Vacunas de Productos Inactivados/administración & dosificación
13.
Clin Sci (Lond) ; 135(17): 2067-2083, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34405230

RESUMEN

Dipeptidyl peptidase 4 (DPP4) expression is increased in the lungs of chronic obstructive pulmonary disease (COPD). DPP4 is known to be associated with inflammation in various organs, including LPS-induced acute lung inflammation. Since non-typeable Haemophilus influenzae (NTHi) causes acute exacerbations in COPD patients, we examined the contribution of DPP4 in NTHi-induced lung inflammation in COPD. Pulmonary macrophages isolated from COPD patients showed higher expression of DPP4 than the macrophages isolated from normal subjects. In response to NTHi infection, COPD, but not normal macrophages show a further increase in the expression of DPP4. COPD macrophages also showed higher expression of IL-1ß, and CCL3 responses to NTHi than normal, and treatment with DPP4 inhibitor, diprotin A attenuated this response. To examine the contribution of DPP4 in NTHi-induced lung inflammation, COPD mice were infected with NTHi, treated with diprotin A or PBS intraperitoneally, and examined for DPP4 expression, lung inflammation, and cytokine expression. Mice with COPD phenotype showed increased expression of DPP4, which increased further following NTHi infection. DPP4 expression was primarily observed in the infiltrated inflammatory cells. NTHi-infected COPD mice also showed sustained neutrophilic lung inflammation and expression of CCL3, and this was inhibited by DPP4 inhibitor. These observations indicate that enhanced expression of DPP4 in pulmonary macrophages may contribute to sustained lung inflammation in COPD following NTHi infection. Therefore, inhibition of DPP4 may reduce the severity of NTHi-induced lung inflammation in COPD.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Infecciones por Haemophilus/enzimología , Haemophilus influenzae/patogenicidad , Macrófagos Alveolares/enzimología , Neumonía Bacteriana/enzimología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Anciano , Animales , Estudios de Casos y Controles , Quimiocina CCL20/metabolismo , Quimiocina CCL3/metabolismo , Modelos Animales de Enfermedad , Femenino , Infecciones por Haemophilus/microbiología , Interacciones Huésped-Patógeno , Humanos , Interleucina-1beta/metabolismo , Macrófagos Alveolares/microbiología , Masculino , Ratones , Persona de Mediana Edad , Neumonía Bacteriana/microbiología , Enfermedad Pulmonar Obstructiva Crónica/microbiología
14.
J Exp Med ; 218(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34292313

RESUMEN

In this study, we detail a novel approach that combines bacterial fitness fluorescent reporter strains with scRNA-seq to simultaneously acquire the host transcriptome, surface marker expression, and bacterial phenotype for each infected cell. This approach facilitates the dissection of the functional heterogeneity of M. tuberculosis-infected alveolar (AMs) and interstitial macrophages (IMs) in vivo. We identify clusters of pro-inflammatory AMs associated with stressed bacteria, in addition to three different populations of IMs with heterogeneous bacterial phenotypes. Finally, we show that the main macrophage populations in the lung are epigenetically constrained in their response to infection, while inter-species comparison reveals that most AMs subsets are conserved between mice and humans. This conceptual approach is readily transferable to other infectious disease agents with the potential for an increased understanding of the roles that different host cell populations play during the course of an infection.


Asunto(s)
Macrófagos Alveolares/microbiología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/genética , Tuberculosis Pulmonar/patología , Animales , Antituberculosos/farmacología , Líquido del Lavado Bronquioalveolar/microbiología , Antígenos CD11/inmunología , Antígenos CD11/metabolismo , Epigénesis Genética , Regulación Bacteriana de la Expresión Génica , Hemo/metabolismo , Interacciones Huésped-Patógeno , Humanos , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Ratones Endogámicos C57BL , Microorganismos Modificados Genéticamente , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/microbiología
15.
Methods Mol Biol ; 2314: 167-182, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34235652

RESUMEN

Mycobacterium tuberculosis colonizes, survives, and grows inside macrophages. In vitro macrophage infection models, using both primary macrophages and cell lines, enable the characterization of the pathogen response to macrophage immune pressure and intracellular environmental cues. We describe methods to propagate and infect primary murine bone marrow-derived macrophages, HoxB8 conditionally immortalized myeloid cells, Max Planck Institute alveolar macrophage-like cells, and J774 and THP-1 macrophage-like cell lines. We also present methods on the characterization of M. tuberculosis intracellular survival and the preparation of infected macrophages for imaging.


Asunto(s)
Macrófagos Alveolares/microbiología , Macrófagos/microbiología , Imagen Molecular/métodos , Mycobacterium tuberculosis/crecimiento & desarrollo , Células Mieloides/microbiología , Animales , Células Cultivadas , Humanos , Técnicas In Vitro , Macrófagos/patología , Macrófagos Alveolares/patología , Ratones , Mycobacterium tuberculosis/patogenicidad , Células Mieloides/patología
16.
mSphere ; 6(3): e0026021, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34077261

RESUMEN

Aspergillus fumigatus is the most common cause of mold pneumonia worldwide, and a significant cause of infectious morbidity and mortality in immunocompromised individuals. The oxidative burst, which generates reactive oxidative species (ROS), plays a pivotal role in host defense against aspergillosis and induces regulated cell death in Aspergillus conidia, the infectious propagules. Beyond the well-established role of NADP (NADPH) oxidase in ROS generation by neutrophils and other innate effector cells, mitochondria represent a major ROS production site in many cell types, though it is unclear whether mitochondrial ROS (mtROS) contribute to antifungal activity in the lung. Following A. fumigatus infection, we observed that innate effector cells, including alveolar macrophages (AMs), monocyte-derived dendritic cells (Mo-DCS), and neutrophils, generated mtROS, primarily in fungus-infected cells. To examine the functional role of mtROS, specifically the H2O2 component, in pulmonary host defense against A. fumigatus, we infected transgenic mice that expressed a mitochondrion-targeted catalase. Using a reporter of fungal viability during interactions with leukocytes, mitochondrial H2O2 (mtH2O2) was essential for optimal AM, but not for neutrophil phagocytic and conidiacidal activity in the lung. Catalase-mediated mtH2O2 neutralization did not lead to invasive aspergillosis in otherwise immunocompetent mice and did not shorten survival in mice that lack NADPH oxidase function. Collectively, these studies indicate that mtROS-associated defects in AM antifungal activity can be functionally compensated by the action of NADPH oxidase and by nonoxidative effector mechanisms during murine A. fumigatus lung infection. IMPORTANCE Aspergillus fumigatus is a fungal pathogen that causes invasive disease in humans with defects in immune function. Airborne conidia, the infectious propagules, are ubiquitous and inhaled on a daily basis. In the respiratory tree, conidia are killed by the coordinated actions of phagocytes, including alveolar macrophages, neutrophils, and monocyte-derived dendritic cells. The oxidative burst represents a central killing mechanism and relies on the assembly of the NADPH oxidase complex on the phagosomal membrane. However, NADPH oxidase-deficient leukocytes have significant residual fungicidal activity in vivo, indicating the presence of alternative effector mechanisms. Here, we report that murine innate immune cells produce mitochondrial reactive oxygen species (mtROS) in response to fungal interactions. Neutralizing the mtROS constituent hydrogen peroxide (H2O2) via a catalase expressed in mitochondria of innate immune cells substantially diminished fungicidal properties of alveolar macrophages, but not of other innate immune cells. These data indicate that mtH2O2 represent a novel AM killing mechanism against Aspergillus conidia. mtH2O2 neutralization is compensated by other killing mechanisms in the lung, demonstrating functional redundancy at the level of host defense in the respiratory tree. These findings have important implications for the development of host-directed therapies against invasive aspergillosis in susceptible patient populations.


Asunto(s)
Aspergillus fumigatus/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Macrófagos Alveolares/inmunología , Mitocondrias/inmunología , Especies Reactivas de Oxígeno/inmunología , Animales , Aspergilosis/inmunología , Aspergillus fumigatus/patogenicidad , Peróxido de Hidrógeno/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo
17.
PLoS Biol ; 19(6): e3001247, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34061822

RESUMEN

Aspergillus fumigatus is a human fungal pathogen that can cause devastating pulmonary infections, termed "aspergilloses," in individuals suffering immune imbalances or underlying lung conditions. As rapid adaptation to stress is crucial for the outcome of the host-pathogen interplay, here we investigated the role of the versatile posttranslational modification (PTM) persulfidation for both fungal virulence and antifungal host defense. We show that an A. fumigatus mutant with low persulfidation levels is more susceptible to host-mediated killing and displays reduced virulence in murine models of infection. Additionally, we found that a single nucleotide polymorphism (SNP) in the human gene encoding cystathionine γ-lyase (CTH) causes a reduction in cellular persulfidation and correlates with a predisposition of hematopoietic stem cell transplant recipients to invasive pulmonary aspergillosis (IPA), as correct levels of persulfidation are required for optimal antifungal activity of recipients' lung resident host cells. Importantly, the levels of host persulfidation determine the levels of fungal persulfidation, ultimately reflecting a host-pathogen functional correlation and highlighting a potential new therapeutic target for the treatment of aspergillosis.


Asunto(s)
Antifúngicos/farmacología , Aspergillus fumigatus/patogenicidad , Proteínas Fúngicas/metabolismo , Interacciones Huésped-Patógeno , Sulfuros/metabolismo , Células A549 , Adulto , Animales , Aspergilosis/epidemiología , Aspergilosis/genética , Aspergilosis/microbiología , Aspergillus fumigatus/efectos de los fármacos , Aspergillus fumigatus/enzimología , Cistationina gamma-Liasa/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Incidencia , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/microbiología , Masculino , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Polimorfismo de Nucleótido Simple/genética , Células THP-1 , Receptores de Trasplantes , Virulencia/efectos de los fármacos , Adulto Joven
18.
Front Immunol ; 12: 629281, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33968022

RESUMEN

Alveolar macrophages (AMs) are pivotal for maintaining lung immune homeostasis. We demonstrated that deletion of liver kinase b1 (Lkb1) in CD11c+ cells led to greatly reduced AM abundance in the lung due to the impaired self-renewal of AMs but not the impeded pre-AM differentiation. Mice with Lkb1-deficient AMs exhibited deteriorated diseases during airway Staphylococcus aureus (S. aureus) infection and allergic inflammation, with excessive accumulation of neutrophils and more severe lung pathology. Drug-mediated AM depletion experiments in wild type mice indicated a cause for AM reduction in aggravated diseases in Lkb1 conditional knockout mice. Transcriptomic sequencing also revealed that Lkb1 inhibited proinflammatory pathways, including IL-17 signaling and neutrophil migration, which might also contribute to the protective function of Lkb1 in AMs. We thus identified Lkb1 as a pivotal regulator that maintains the self-renewal and immune function of AMs.


Asunto(s)
Asma/enzimología , Autorrenovación de las Células , Pulmón/enzimología , Macrófagos Alveolares/enzimología , Neumonía Bacteriana/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Infecciones Estafilocócicas/enzimología , Proteínas Quinasas Activadas por AMP , Animales , Asma/genética , Asma/inmunología , Antígenos CD11/genética , Antígenos CD11/metabolismo , Modelos Animales de Enfermedad , Homeostasis , Interleucina-17/genética , Interleucina-17/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Neumonía Bacteriana/genética , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Transcriptoma
19.
Front Immunol ; 12: 635097, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33968026

RESUMEN

In Glässer's disease outbreaks, Glaesserella (Haemophilus) parasuis has to overcome the non-specific immune system in the lower respiratory tract, the alveolar macrophages. Here we showed that porcine alveolar macrophages (PAMs) were able to recognize and phagocyte G. parasuis with strain-to-strain variability despite the presence of the capsule in virulent (serovar 1, 5, 12) as well in avirulent strains (serovar 6 and 9). The capsule, outer membrane proteins, virulence-associated autotransporters, cytolethal distending toxins and many other proteins have been identified as virulence factors of this bacterium. Therefore, we immunized pigs with the crude capsular extract (cCE) from the virulent G. parasuis CAPM 6475 strain (serovar 5) and evaluated the role of the anti-cCE/post-vaccinal IgG in the immune response of PAMs to in vitro infection with various G. parasuis strains. We demonstrated the specific binding of the antibodies to the cCE by Western-blotting assay and immunoprecipitation as well as the specific binding to the strain CAPM 6475 in transmission electron microscopy. In the cCE, we identified several virulence-associated proteins that were immunoreactive with IgG isolated from sera of immunized pigs. Opsonization of G. parasuis strains by post-vaccinal IgG led to enhanced phagocytosis of G. parasuis by PAMs at the first two hours of infection. Moreover, opsonization increased the oxidative burst and expression/production of both pro- and anti-inflammatory cytokines. The neutralizing effects of these antibodies on the antioxidant mechanisms of G. parasuis may lead to attenuation of its virulence and pathogenicity in vivo. Together with opsonization of bacteria by these antibodies, the host may eliminate G. parasuis in the infection site more efficiently. Based on these results, the crude capsular extract is a vaccine candidate with immunogenic properties.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Cápsulas Bacterianas/inmunología , Infecciones por Haemophilus/inmunología , Haemophilus parasuis/inmunología , Macrófagos Alveolares/inmunología , Animales , Anticuerpos Antibacterianos/metabolismo , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/metabolismo , Especificidad de Anticuerpos , Células Cultivadas , Infecciones por Haemophilus/metabolismo , Infecciones por Haemophilus/microbiología , Haemophilus parasuis/patogenicidad , Cinética , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiología , Fagocitosis , Especies Reactivas de Oxígeno/metabolismo , Serogrupo , Sus scrofa , Virulencia
20.
Respir Res ; 22(1): 159, 2021 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-34022899

RESUMEN

BACKGROUND: Patients in intensive care units (ICUs) often received broad-spectrum antibiotic treatment and Acinetobacter baumannii (A.b.) and Pseudomonas aeruginosa (P.a.) were the most common pathogens causing ventilator-associated pneumonia (VAP). This study aimed to examine the effects and mechanism of mechanical ventilation (MV) on A.b.-induced lung injury and the involvement of alveolar macrophages (AMs). METHODS: C57BL/6 wild-type (WT) and c-Jun N-terminal kinase knockout (JNK1-/-) mice received MV for 3 h at 2 days after nasal instillation of A.b., P.a. (1 × 106 colony-forming unit, CFU), or normal saline. RESULTS: Intranasal instillation of 106 CFU A.b. in C57BL/6 mice induced a significant increase in total cells and protein levels in the bronchoalveolar lavage fluid (BALF) and neutrophil infiltration in the lungs. MV after A.b. instillation increases neutrophil infiltration, interleukin (IL)-6 and vascular cell adhesion molecule (VCAM) mRNA expression in the lungs and total cells, IL-6 levels, and nitrite levels in the BALF. The killing activity of AMs against A.b. was lower than against P.a. The diminished killing activity was parallel with decreased tumor necrosis factor-α production by AMs compared with A.b. Inducible nitric oxide synthase inhibitor, S-methylisothiourea, decreased the total cell number in BALF on mice receiving A.b. instillation and ventilation. Moreover, MV decreased the A.b. and P.a. killing activity of AMs. MV after A.b. instillation induced less total cells in the BALF and nitrite production in the serum of JNK1-/- mice than those of WT mice. CONCLUSION: A.b. is potent in inducing neutrophil infiltration in the lungs and total protein in the BALF. MV enhances A.b.-induced lung injury through an increase in the expression of VCAM and IL-6 levels in the BALF and a decrease in the bacteria-killing activity of AMs. A lower inflammation level in JNK1-/- mice indicates that A.b.-induced VAP causes lung injury through JNK signaling pathway in the lungs.


Asunto(s)
Infecciones por Acinetobacter/enzimología , Acinetobacter baumannii/patogenicidad , Pulmón/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Neumonía Asociada al Ventilador/enzimología , Respiración Artificial/efectos adversos , Lesión Pulmonar Inducida por Ventilación Mecánica/enzimología , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/genética , Infiltración Neutrófila , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neumonía Asociada al Ventilador/microbiología , Neumonía Asociada al Ventilador/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/microbiología , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...