Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67.497
Filtrar
1.
Zhonghua Er Ke Za Zhi ; 62(7): 669-675, 2024 Jul 02.
Artículo en Chino | MEDLINE | ID: mdl-38955686

RESUMEN

Objective: To investigate and summarize pediatric patients with severe Mycoplasma pneumoniae pneumonia (MPP) presenting with varied clinical and chest imaging features in order to guide the individualized treatment. Methods: This was a retrospective cohort study. Medical records of clinical, imaging and laboratory data of 505 patients with MPP who were admitted to the Department Ⅱ of Respirology Center, Beijing Children's Hospital, Capital Medical University from January 2016 to October 2023 and met the enrollment criteria were included. They were divided into severe group and non-severe group according to whether lower airway obliterans was developed. The clinical and chest imaging features of the two groups were analyzed. Those severe cases with single lobe ≥2/3 consolidation (lobar consolidation) were further divided into subtype lung-necrosis and subtype non-lung-necrosis based on whether lung necrosis was developed. Comparison on the clinical manifestations, bronchoscopic findings, whole blood C-reactive protein (CRP) and other inflammatory indicators between the two subtypes was performed. Comparisons between two groups were achieved using independent-sample t-test, nonparametric test or chi-square test. Univariate receiver operating characteristic (ROC) curve analyses were performed on the indicators such as CRP of the two subtypes. Results: Of the 505 cases, 254 were male and 251 were female. The age of the onset was (8.2±2.9) years. There were 233 severe cases, among whom 206 were with lobar consolidation and 27 with diffuse bronchiolitis. The other 272 belonged to non-severe cases, with patchy, cloudy infiltrations or single lobe <2/3 uneven consolidation or localized bronchiolitis. Of the 206 cases (88.4%) severe cases with lobar consolidation, 88 harbored subtype lung-necrosis and 118 harbored subtype non-lung-necrosis. All 206 cases (100.0%) presented with persistent high fever, among whom 203 cases (98.5%) presented with inflammatory secretion obstruction and plastic bronchitis under bronchoscopy. Of those 88 cases with subtype lung-necrosis, there were 42 cases (47.7%) with dyspnea and 39 cases (44.3%) with moderate to massive amount of pleural effusion. There were 35 cases (39.8%) diagnosed with lung embolism during the disease course, of which other 34 cases (38.6%) were highly suspected. Extensive airway mucosal necrosis was observed in 46 cases (52.3%), and the level of their whole blood CRP was significantly higher than that of subtype non-lung-necrosis (131.5 (91.0, 180.0) vs. 25.5 (12.0, 43.1) mg/L, U=334.00, P<0.001). They were regarded as subtype "lung consolidation-atelectasis-necrosis". Of those 118 cases with subtype non-lung-necrosis, 27 cases (22.9%) presented with dyspnea and none were with moderate to massive amount of pleural effusion. Sixty-five cases (55.1%) presented with plastic bronchitis and localized airway mucosal necrosis was observed in 32 cases (27.1%). They were deemed as subtype "lung consolidation-atelectasis". ROC curve analyses revealed that whole blood CRP of 67.5 mg/L on the 6-10 th day of disease course exhibited a sensitivity of 0.96, a specificity of 0.89, and an area under the curve of 0.97 for distinguishing between these two subtypes among those with lobar consolidation. Conclusions: Pediatric patients with severe MPP present with lobar consolidation or diffuse bronchiolitis on chest imaging. Those with lobar consolidation harbor 2 subtypes as "lung consolidation-atelectasis-necrosis" and "lung consolidation-atelectasis". Whole blood CRP of 67.5 mg/L can be applied as an early discriminating indicator to discriminate between these two subtypes.


Asunto(s)
Proteína C-Reactiva , Pulmón , Mycoplasma pneumoniae , Fenotipo , Neumonía por Mycoplasma , Humanos , Femenino , Masculino , Neumonía por Mycoplasma/diagnóstico , Estudios Retrospectivos , Niño , Pulmón/patología , Pulmón/diagnóstico por imagen , Proteína C-Reactiva/análisis , Broncoscopía/métodos , Índice de Severidad de la Enfermedad , Preescolar , Necrosis , Bronquiolitis/diagnóstico , Bronquiolitis/patología
3.
Front Immunol ; 15: 1379570, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38957465

RESUMEN

There is a reciprocal relationship between extracellular matrix (ECM) remodelling and inflammation that could be operating in the progression of severe COVID-19. To explore the immune-driven ECM remodelling in COVID-19, we in this explorative study analysed these interactions in hospitalised COVID-19 patients. RNA sequencing and flow analysis were performed on peripheral blood mononuclear cells. Inflammatory mediators in plasma were measured by ELISA and MSD, and clinical information from hospitalised COVID-19 patients (N=15) at admission was included in the analysis. Further, we reanalysed two publicly available datasets: (1) lung tissue RNA-sequencing dataset (N=5) and (2) proteomics dataset from PBCM. ECM remodelling pathways were enriched in PBMC from COVID-19 patients compared to healthy controls. Patients treated at the intensive care unit (ICU) expressed distinct ECM remodelling gene profiles compared to patients in the hospital ward. Several markers were strongly correlated to immune cell subsets, and the dysregulation in the ICU patients was positively associated with plasma levels of inflammatory cytokines and negatively associated with B-cell activating factors. Finally, our analysis of publicly accessible datasets revealed (i) an augmented ECM remodelling signature in inflamed lung tissue compared to non-inflamed tissue and (ii) proteomics analysis of PBMC from severe COVID-19 patients demonstrated an up-regulation in an ECM remodelling pathway. Our results may suggest the presence of an interaction between ECM remodelling, inflammation, and immune cells, potentially initiating or perpetuating pulmonary pathology in severe COVID-19.


Asunto(s)
COVID-19 , Matriz Extracelular , Leucocitos Mononucleares , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/sangre , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Matriz Extracelular/metabolismo , Masculino , Femenino , Persona de Mediana Edad , SARS-CoV-2/fisiología , SARS-CoV-2/inmunología , Anciano , Citocinas/sangre , Proteómica/métodos , Pulmón/inmunología , Pulmón/patología , Adulto
4.
Sci Transl Med ; 16(754): eadi6887, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38959328

RESUMEN

Virulent infectious agents such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and methicillin-resistant Staphylococcus aureus (MRSA) induce tissue damage that recruits neutrophils, monocyte, and macrophages, leading to T cell exhaustion, fibrosis, vascular leak, epithelial cell depletion, and fatal organ damage. Neutrophils, monocytes, and macrophages recruited to pathogen-infected lungs, including SARS-CoV-2-infected lungs, express phosphatidylinositol 3-kinase gamma (PI3Kγ), a signaling protein that coordinates both granulocyte and monocyte trafficking to diseased tissues and immune-suppressive, profibrotic transcription in myeloid cells. PI3Kγ deletion and inhibition with the clinical PI3Kγ inhibitor eganelisib promoted survival in models of infectious diseases, including SARS-CoV-2 and MRSA, by suppressing inflammation, vascular leak, organ damage, and cytokine storm. These results demonstrate essential roles for PI3Kγ in inflammatory lung disease and support the potential use of PI3Kγ inhibitors to suppress inflammation in severe infectious diseases.


Asunto(s)
COVID-19 , Fosfatidilinositol 3-Quinasa Clase Ib , Inflamación , SARS-CoV-2 , COVID-19/patología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Animales , Inflamación/patología , Humanos , Tratamiento Farmacológico de COVID-19 , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Pulmón/patología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Permeabilidad Capilar/efectos de los fármacos , Ratones Endogámicos C57BL , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/patología
5.
Georgian Med News ; (349): 54-59, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38963202

RESUMEN

Doxorubicin is the common chemotherapeutic agent that has been harnessed for the treatment of various types of malignancy including the treatment of soft tissue and osteosarcoma and cancers of the vital organs like breast, ovary, bladder, and thyroid. It is also used to treat leukaemia and lymphoma, however, this is an obstacle because of their prominent side effects including cardiotoxicity and lung fibrosis, we do aim to determine the role of CoQ10 as an antioxidant on the impeding the deleterious impacts of doxorubicin on tissue degenerative effects. To do so, 27 rats were subdivided into 3 groups of 9 each; CoQ10 exposed group, Doxorubicin exposed group, and CoQ10 plus Doxorubicin group. At the end of the study, the animals were sacrificed and lungs with hearts were harvested, and slides were prepared for examination under a microscope. The results indicated that doxorubicin induced abnormal cellular structure resulting in damaging cellular structures of the lung and heart while CoQ10 impeded these damaging effects and nearly restoring normal tissue structure. As a result, CoQ10 will maintain normal tissue of the lung and heart.


Asunto(s)
Doxorrubicina , Pulmón , Ubiquinona , Animales , Doxorrubicina/efectos adversos , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Ratas , Pulmón/efectos de los fármacos , Pulmón/patología , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/toxicidad , Miocardio/patología , Masculino , Antioxidantes/farmacología , Cardiotoxicidad/etiología , Cardiotoxicidad/patología , Corazón/efectos de los fármacos
6.
BMC Pulm Med ; 24(1): 313, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961422

RESUMEN

BACKGROUND: Primary pulmonary myxoid sarcoma (PPMS) is a rare, low-grade malignant tumor, constituting approximately 0.2% of all lung tumors. Despite its rarity, PPMS possesses distinctive histological features and molecular alterations, notably the presence of EWSR1-CREB1 gene fusion. However, its precise tissue origin remains elusive, posing challenges in clinical diagnosis. CASE DEMONSTRATION: A 20-year-old male patient underwent a routine physical examination 6 months prior, revealing a pulmonary mass. Following surgical excision, microscopic evaluation unveiled predominantly short spindle-shaped tumor cells organized in a fascicular, beam-like, or reticular pattern. The stromal matrix exhibited abundant mucin, accompanied by lymphocytic and plasma cell infiltration, with Russell bodies evident in focal areas. Immunophenotypic profiling revealed positive expression of vimentin and epithelial membrane antigen in tumor cells, whereas smooth muscle actin and S-100, among others, were negative. Ki-67 proliferation index was approximately 5%. Subsequent second-generation sequencing identified the characteristic EWSR1-CREB1 gene fusion. The definitive pathological diagnosis established PPMS. The patient underwent no adjuvant chemotherapy or radiotherapy and remained recurrence-free during a 30-month follow-up period. CONCLUSIONS: We report a rare case of PPMS located within the left lung lobe interlobar fissure, featuring Russell body formation within the tumor stroma, a novel finding in PPMS. Furthermore, the histomorphological characteristics of this case highlight the diagnostic challenge it poses, as it may mimic inflammatory myofibroblastic tumor, extraskeletal myxoid chondrosarcoma, or hemangiopericytoma-like fibrous histiocytoma. Therefore, accurate diagnosis necessitates an integrated approach involving morphological, immunohistochemical, and molecular analyses.


Asunto(s)
Neoplasias Pulmonares , Humanos , Masculino , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/diagnóstico , Adulto Joven , Proteínas de Fusión Oncogénica/genética , Tomografía Computarizada por Rayos X , Mixosarcoma/patología , Mixosarcoma/genética , Mixosarcoma/cirugía , Mixosarcoma/diagnóstico , Sarcoma/genética , Sarcoma/patología , Sarcoma/diagnóstico , Sarcoma/cirugía , Pulmón/patología , Pulmón/diagnóstico por imagen
7.
J Transl Med ; 22(1): 615, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961500

RESUMEN

OBJECTIVE: To explore the correlation between asthma risk and genetic variants affecting the expression or function of lipid-lowering drug targets. METHODS: We conducted Mendelian randomization (MR) analyses using variants in several genes associated with lipid-lowering medication targets: HMGCR (statin target), PCSK9 (alirocumab target), NPC1L1 (ezetimibe target), APOB (mipomersen target), ANGPTL3 (evinacumab target), PPARA (fenofibrate target), and APOC3 (volanesorsen target), as well as LDLR and LPL. Our objective was to investigate the relationship between lipid-lowering drugs and asthma through MR. Finally, we assessed the efficacy and stability of the MR analysis using the MR Egger and inverse variance weighted (IVW) methods. RESULTS: The elevated triglyceride (TG) levels associated with the APOC3, and LPL targets were found to increase asthma risk. Conversely, higher LDL-C levels driven by LDLR were found to decrease asthma risk. Additionally, LDL-C levels (driven by APOB, NPC1L1 and HMGCR targets) and TG levels (driven by the LPL target) were associated with improved lung function (FEV1/FVC). LDL-C levels driven by PCSK9 were associated with decreased lung function (FEV1/FVC). CONCLUSION: In conclusion, our findings suggest a likely causal relationship between asthma and lipid-lowering drugs. Moreover, there is compelling evidence indicating that lipid-lowering therapies could play a crucial role in the future management of asthma.


Asunto(s)
Asma , Hipolipemiantes , Análisis de la Aleatorización Mendeliana , Humanos , Asma/genética , Asma/tratamiento farmacológico , Hipolipemiantes/uso terapéutico , Hipolipemiantes/farmacología , Proproteína Convertasa 9/genética , Estudios de Asociación Genética , Pulmón/efectos de los fármacos , Pulmón/patología , Lipoproteína Lipasa/genética , Triglicéridos/sangre , Receptores de LDL/genética , Hidroximetilglutaril-CoA Reductasas/genética , Proteína 3 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina/genética , Apolipoproteína C-III/genética , Apolipoproteínas B/genética , Pruebas de Función Respiratoria , LDL-Colesterol/sangre , Proteínas de Transporte de Membrana , PPAR alfa
8.
Front Immunol ; 15: 1424374, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38966641

RESUMEN

At the beginning of the COVID-19 pandemic those with underlying chronic lung conditions, including tuberculosis (TB), were hypothesized to be at higher risk of severe COVID-19 disease. However, there is inconclusive clinical and preclinical data to confirm the specific risk SARS-CoV-2 poses for the millions of individuals infected with Mycobacterium tuberculosis (M.tb). We and others have found that compared to singly infected mice, mice co-infected with M.tb and SARS-CoV-2 leads to reduced SARS-CoV-2 severity compared to mice infected with SARS-CoV-2 alone. Consequently, there is a large interest in identifying the molecular mechanisms responsible for the reduced SARS-CoV-2 infection severity observed in M.tb and SARS-CoV-2 co-infection. To address this, we conducted a comprehensive characterization of a co-infection model and performed mechanistic in vitro modeling to dynamically assess how the innate immune response induced by M.tb restricts viral replication. Our study has successfully identified several cytokines that induce the upregulation of anti-viral genes in lung epithelial cells, thereby providing protection prior to challenge with SARS-CoV-2. In conclusion, our study offers a comprehensive understanding of the key pathways induced by an existing bacterial infection that effectively restricts SARS-CoV-2 activity and identifies candidate therapeutic targets for SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Coinfección , Inmunidad Innata , Mycobacterium tuberculosis , SARS-CoV-2 , COVID-19/inmunología , Animales , Mycobacterium tuberculosis/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Ratones , Coinfección/inmunología , Humanos , Tuberculosis/inmunología , Tuberculosis/microbiología , Citocinas/metabolismo , Citocinas/inmunología , Modelos Animales de Enfermedad , Índice de Severidad de la Enfermedad , Pulmón/inmunología , Pulmón/virología , Pulmón/microbiología , Pulmón/patología , Replicación Viral , Ratones Endogámicos C57BL , Femenino
9.
Virol J ; 21(1): 151, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965616

RESUMEN

BACKGROUND: The canine influenza virus (CIV) outbreak has garnered considerable attention as it poses a significant threat to dog health. During the H3N2 CIV evolution in beagles, the virus formed a new clade after 2019 and gradually became more adaptable to other mammals. Therefore, successfully elucidating the biological characteristics and constructing a canine influenza infection model is required for CIV characterization. METHODS: We performed genetic analyses to examine the biological characteristics and infection dynamics of CIV. RESULTS: The genotype of our H3N2 CIV strain (from 2019 in Shanghai) belonged to the 5.1 clade, which is now prevalent in China. Using MDCK cells, we investigated viral cytopathic effects. Virus size and morphology were observed using transmission electron microscopy. Beagles were also infected with 104, 105, and 106 50% egg-infectious doses (EID50). When compared with the other groups, the 106 EID50 group showed the most obvious clinical symptoms, the highest virus titers, and typical lung pathological changes. Our results suggested that the other two treatments caused mild clinical manifestations and pathological changes. Subsequently, CIV distribution in the 106 EID50 group was detected by hematoxylin and eosin (H&E) and immunofluorescence (IF) staining, which indicated that CIV primarily infected the lungs. CONCLUSIONS: The framework established in this study will guide further CIV prevention strategies.


Asunto(s)
Enfermedades de los Perros , Genotipo , Subtipo H3N2 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Animales , Perros , Subtipo H3N2 del Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/patología , Enfermedades de los Perros/virología , Células de Riñón Canino Madin Darby , China/epidemiología , Pulmón/virología , Pulmón/patología , Filogenia , Carga Viral , Modelos Animales de Enfermedad
10.
Allergol Immunopathol (Madr) ; 52(4): 15-20, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38970260

RESUMEN

BACKGROUND: Pulmonary fibrosis (PF) is a chronic, progressive, and irreversible heterogeneous disease of lung interstitial tissue. To combat progression of PF, new drugs are required to be developed. Rhizoma coptidis (COP), one of the main alkaloids of Coptis chinensis, is a traditional herbal medicine used to treat various inflammatory diseases. OBJECTIVE: To investigate the possible effects of Coptisine (Cop) on the growth, inflammation, as well as FMT of TNF-ß1-induced HFL1 cells and uncover the mechanism. MATERIAL AND METHODS: Human fetal lung fibroblast 1 (HFL1) was induced using 6ng/mL TGF-ß1 as a model of pulmonary fibrosis. CCK-8, Brdu, and transwell assays indicated the effects on cell growth as well as motility. qPCR and the corresponding kits indicted the effects on cell inflammation. Immunoblot showed the effects on FMT and further confirmed the mechanism. RESULTS: Coptisine inhibits excessive growth as well as motility of TNF-ß1-induced HFL1 cells. It further inhibits inflammation and ROS levels in TNF-ß1-induced HFL1 cells. Coptisine inhibits the FMT process of TNF-ß1-induced HFL1 cells. Mechanically, coptisine promotes the Nrf2/HO-1 pathway. CONCLUSION: Coptisine can inhibit the excessive growth, inflammation as well as FMT of lung fibroblasts into myofibroblasts. It could serve as a promising drug of PF.


Asunto(s)
Berberina , Proliferación Celular , Fibroblastos , Pulmón , Miofibroblastos , Humanos , Proliferación Celular/efectos de los fármacos , Berberina/farmacología , Berberina/análogos & derivados , Miofibroblastos/efectos de los fármacos , Pulmón/patología , Pulmón/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Inflamación/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Factor de Crecimiento Transformador beta1/metabolismo , Línea Celular , Coptis , Hemo-Oxigenasa 1/metabolismo , Transducción de Señal/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Diferenciación Celular/efectos de los fármacos , Antiinflamatorios/farmacología
11.
Cell Mol Life Sci ; 81(1): 287, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38970705

RESUMEN

Lung type 2 pneumocytes (T2Ps) and alveolar macrophages (AMs) play crucial roles in the synthesis, recycling and catabolism of surfactant material, a lipid/protein fluid essential for respiratory function. The liver X receptors (LXR), LXRα and LXRß, are transcription factors important for lipid metabolism and inflammation. While LXR activation exerts anti-inflammatory actions in lung injury caused by lipopolysaccharide (LPS) and other inflammatory stimuli, the full extent of the endogenous LXR transcriptional activity in pulmonary homeostasis is incompletely understood. Here, using mice lacking LXRα and LXRß as experimental models, we describe how the loss of LXRs causes pulmonary lipidosis, pulmonary congestion, fibrosis and chronic inflammation due to defective de novo synthesis and recycling of surfactant material by T2Ps and defective phagocytosis and degradation of excess surfactant by AMs. LXR-deficient T2Ps display aberrant lamellar bodies and decreased expression of genes encoding for surfactant proteins and enzymes involved in cholesterol, fatty acids, and phospholipid metabolism. Moreover, LXR-deficient lungs accumulate foamy AMs with aberrant expression of cholesterol and phospholipid metabolism genes. Using a house dust mite aeroallergen-induced mouse model of asthma, we show that LXR-deficient mice exhibit a more pronounced airway reactivity to a methacholine challenge and greater pulmonary infiltration, indicating an altered physiology of LXR-deficient lungs. Moreover, pretreatment with LXR agonists ameliorated the airway reactivity in WT mice sensitized to house dust mite extracts, confirming that LXR plays an important role in lung physiology and suggesting that agonist pharmacology could be used to treat inflammatory lung diseases.


Asunto(s)
Homeostasis , Receptores X del Hígado , Macrófagos Alveolares , Neumonía , Surfactantes Pulmonares , Transducción de Señal , Animales , Receptores X del Hígado/metabolismo , Receptores X del Hígado/genética , Surfactantes Pulmonares/metabolismo , Ratones , Neumonía/metabolismo , Neumonía/patología , Macrófagos Alveolares/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Pulmón/metabolismo , Pulmón/patología , Células Epiteliales Alveolares/metabolismo , Asma/metabolismo , Asma/patología , Asma/genética , Colesterol/metabolismo , Metabolismo de los Lípidos , Fagocitosis
12.
Adv Exp Med Biol ; 1445: 157-168, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38967758

RESUMEN

As the locus for air exchange, lung tissue is perpetually exposed to a significant quantity of foreign pathogens. Consequently, lung has developed a refined and intricate immune system. Beyond their physical and chemical barrier roles, lung epithelial cells can contribute to immune defence through the expression of Toll-like receptors (TLRs) and other pattern recognition receptors, along with the secretion of cytokines. Emerging evidence demonstrates that lung epithelial cells can generate and secrete immunoglobulins (Igs), including IgM, IgA, or IgG, thus performing antibody function. Moreover, malignantly transformed lung epithelial cells have been discovered to produce high levels of Ig, predominantly IgG, which do not fulfill the role of antibodies, but instead carries out tumour-promoting activity. Structural analysis has indicated that the biological activity of IgG produced by lung cancer cells differs from that of Igs produced by normal lung epithelial cells due to the unique glycosylation modification. Specifically, the sialylated IgG (SIA-IgG), characterised by a non-traditional N-glycosylation modification at the Asn162 site of Igγ CH1, is highly expressed in tumour stem cells. It has been demonstrated that SIA-IgG relies on this unique sialylation modification to promote tumorigenesis, metastasis, and immune evasion. Current results have proven that the Ig produced by lung epithelial cells has multifaceted biological activities, including immune defence functions under physiological conditions, while acquiring tumour-promoting activity during malignant transformation. These insights possess potential for the diagnosis and treatment of lung cancer as novel biomarkers and targets.


Asunto(s)
Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Animales , Células Epiteliales/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/patología , Glicosilación , Pulmón/inmunología , Pulmón/patología , Pulmón/metabolismo , Inmunoglobulinas/metabolismo , Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo
13.
J Med Primatol ; 53(4): e12722, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38949157

RESUMEN

BACKGROUND: Tuberculosis (TB) kills approximately 1.6 million people yearly despite the fact anti-TB drugs are generally curative. Therefore, TB-case detection and monitoring of therapy, need a comprehensive approach. Automated radiological analysis, combined with clinical, microbiological, and immunological data, by machine learning (ML), can help achieve it. METHODS: Six rhesus macaques were experimentally inoculated with pathogenic Mycobacterium tuberculosis in the lung. Data, including Computed Tomography (CT), were collected at 0, 2, 4, 8, 12, 16, and 20 weeks. RESULTS: Our ML-based CT analysis (TB-Net) efficiently and accurately analyzed disease progression, performing better than standard deep learning model (LLM OpenAI's CLIP Vi4). TB-Net based results were more consistent than, and confirmed independently by, blinded manual disease scoring by two radiologists and exhibited strong correlations with blood biomarkers, TB-lesion volumes, and disease-signs during disease pathogenesis. CONCLUSION: The proposed approach is valuable in early disease detection, monitoring efficacy of therapy, and clinical decision making.


Asunto(s)
Biomarcadores , Aprendizaje Profundo , Macaca mulatta , Mycobacterium tuberculosis , Tomografía Computarizada por Rayos X , Animales , Biomarcadores/sangre , Tomografía Computarizada por Rayos X/veterinaria , Tuberculosis/veterinaria , Tuberculosis/diagnóstico por imagen , Modelos Animales de Enfermedad , Tuberculosis Pulmonar/diagnóstico por imagen , Masculino , Femenino , Pulmón/diagnóstico por imagen , Pulmón/patología , Pulmón/microbiología , Enfermedades de los Monos/diagnóstico por imagen , Enfermedades de los Monos/microbiología
15.
FASEB J ; 38(13): e23756, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38949649

RESUMEN

Asthma is a chronic pulmonary disease with the worldwide prevalence. The structural alterations of airway walls, termed as "airway remodeling", are documented as the core contributor to the airway dysfunction during chronic asthma. Forkhead box transcription factor FOXK2 is a critical regulator of glycolysis, a metabolic reprogramming pathway linked to pulmonary fibrosis. However, the role of FOXK2 in asthma waits further explored. In this study, the chronic asthmatic mice were induced via ovalbumin (OVA) sensitization and repetitive OVA challenge. FOXK2 was upregulated in the lungs of OVA mice and downregulated after adenovirus-mediated FOXK2 silencing. The lung inflammation, peribronchial collagen deposition, and glycolysis in OVA mice were obviously attenuated after FOXK2 knockdown. Besides, the expressions of FOXK2 and SIRT2 in human bronchial epithelial cells (BEAS-2B) were increasingly upregulated upon TGF-ß1 stimulation and downregulated after FOXK2 knockdown. Moreover, the functional loss of FOXK2 remarkably suppressed TGF-ß1-induced epithelial-mesenchymal transition (EMT) and glycolysis in BEAS-2B cells, as manifested by the altered expressions of EMT markers and glycolysis enzymes. The glycolysis inhibitor 2-deoxy-d-glucose (2-DG) inhibited the EMT in TGF-ß1-induced cells, making glycolysis a driver of EMT. The binding of FOXK2 to SIRT2 was validated, and SIRT2 overexpression blocked the FOXK2 knockdown-mediated inhibition of EMT and glycolysis in TGF-ß1-treated cells, which suggests that FOXK2 regulates EMT and glycolysis in TGF-ß1-treated cells in a SIRT2-dependnet manner. Collectively, this study highlights the protective effect of FOXK2 knockdown on airway remodeling during chronic asthma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma , Factores de Transcripción Forkhead , Glucólisis , Sirtuina 2 , Asma/metabolismo , Asma/patología , Animales , Sirtuina 2/metabolismo , Sirtuina 2/genética , Ratones , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Humanos , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Transición Epitelial-Mesenquimal , Ratones Endogámicos BALB C , Femenino , Factor de Crecimiento Transformador beta1/metabolismo , Pulmón/metabolismo , Pulmón/patología , Línea Celular
16.
Front Immunol ; 15: 1404122, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38979411

RESUMEN

Introduction: Chronic obstructive pulmonary disease (COPD) is a major global cause of mortality with limited effective treatments. Sirtuins (SIRT) are histone deacetylases that are involved in the regulation of redox and inflammatory homeostasis. Hence, the present study aims to investigate the role of SIRT-2 in modulating inflammation in a murine model of COPD. Methods: COPD in mice was established by cigarette smoke (CS) exposure for 60 days, and AK-7 was used as the specific SIRT-2 inhibitor. AK-7 (100 µg/kg and 200 µg/kg body weight) was administered intranasally 1 h before CS exposure. Molecular docking was performed to analyze the binding affinity of different inflammatory proteins with AK-7. Results: Immune cell analysis showed a significantly increased number of macrophages (F4/80), neutrophils (Gr-1), and lymphocytes (CD4+, CD8+, and CD19+) in the COPD, group and their population was declined by AK-7 administration. Total reactive oxygen species, total inducible nitric oxide synthase, inflammatory mediators such as neutrophil elastase, C-reactive protein, histamine, and cytokines as IL4, IL-6, IL-17, and TNF-α were elevated in COPD and declined in the AK-7 group. However, IL-10 showed reverse results representing anti-inflammatory potency. AK-7 administration by inhibiting SIRT-2 decreased the expression of p-NF-κB, p-P38, p-Erk, and p-JNK and increased the expression of Nrf-2. Furthermore, AK-7 also declined the lung injury by inhibiting inflammation, parenchymal destruction, emphysema, collagen, club cells, and Kohn pores. AK-7 also showed good binding affinity with inflammatory proteins. Discussion: The current study reveals that SIRT-2 inhibition mitigates COPD severity and enhances pulmonary therapeutic interventions, suggesting AK-7 as a potential therapeutic molecule for COPD medication development.


Asunto(s)
FN-kappa B , Estrés Oxidativo , Enfermedad Pulmonar Obstructiva Crónica , Sirtuina 2 , Animales , Sirtuina 2/metabolismo , Sirtuina 2/antagonistas & inhibidores , Ratones , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Estrés Oxidativo/efectos de los fármacos , FN-kappa B/metabolismo , Masculino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Pulmón/patología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Modelos Animales de Enfermedad , Transducción de Señal , Ratones Endogámicos C57BL , Citocinas/metabolismo , Carbazoles
17.
Autoimmunity ; 57(1): 2370536, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38976509

RESUMEN

Lupus, a systemic autoimmune disease shaped by gene-environment interplay, often progresses to endstage renal failure. While subchronic systemic exposure to bacterial lipopolysaccharide (LPS) triggers autoimmunity and glomerulonephritis in lupus-prone mice, it is unknown if inhaling LPS, which is common in certain occupations, can similarly trigger lupus. Here we determined how subchronic intranasal (IN) LPS instillation influences autoimmunity and glomerulonephritis development in lupusprone NZBWF1 female mice. Briefly, mice were IN-instilled with vehicle or E. coli LPS (0.8 µg/g) twice weekly for 5 wk, followed by necropsy. For systemic comparison, additional cohorts of mice were injected with LPS intraperitoneally (IP) using identical doses/timing. Lungs were assessed for inflammatory and autoimmune responses and then related to systemic autoimmunity and glomerulonephritis. IN/LPS exposure induced in the lung: i) leukocyte infiltration, ii)mRNA signatures for cytokines, chemokines, IFN-regulated, and cell death-related genes, iii) ectopic lymphoid tissue formation, and iv)diverse IgM and IgG autoantibodies (AAbs). Pulmonary effects coincided with enlarged spleens, elevated plasma IgG AAbs, and inflamed IgG-containing kidney glomeruli. In contrast, IP/LPS treatment induced systemic autoimmunity and glomerulonephritis without pulmonary manifestations. Taken together, these preclinical findings suggest the lung could serve as a critical nexus for triggering autoimmunity by respirable LPS in genetically predisposed individuals.


Asunto(s)
Administración Intranasal , Autoanticuerpos , Autoinmunidad , Modelos Animales de Enfermedad , Glomerulonefritis , Lipopolisacáridos , Pulmón , Animales , Lipopolisacáridos/inmunología , Ratones , Autoinmunidad/efectos de los fármacos , Glomerulonefritis/inmunología , Glomerulonefritis/inducido químicamente , Glomerulonefritis/etiología , Glomerulonefritis/patología , Femenino , Pulmón/inmunología , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Autoanticuerpos/inmunología , Autoanticuerpos/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/etiología , Citocinas/metabolismo
18.
J Med Virol ; 96(7): e29768, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38978388

RESUMEN

The vagus nerve circuit, operating through the alpha-7 nicotinic acetylcholine receptor (α7 nAChR), regulates the inflammatory response by influencing immune cells. However, the role of vagal-α7 nAChR signaling in influenza virus infection is unclear. In particular, does vagal-α7 nAChR signaling impact the infection of alveolar epithelial cells (AECs), the primary target cells of influenza virus? Here, we demonstrated a distinct role of α7 nAChR in type II AECs compared to its role in immune cells during influenza infection. We found that deletion of Chrna7 (encoding gene of α7 nAChR) in type II AECs or disruption of vagal circuits reduced lung influenza infection and protected mice from influenza-induced lung injury. We further unveiled that activation of α7 nAChR enhanced influenza infection through PTP1B-NEDD4L-ASK1-p38MAPK pathway. Mechanistically, activation of α7 nAChR signaling decreased p38MAPK phosphorylation during infection, facilitating the nuclear export of influenza viral ribonucleoproteins and thereby promoting infection. Taken together, our findings reveal a mechanism mediated by vagal-α7 nAChR signaling that promotes influenza viral infection and exacerbates disease severity. Targeting vagal-α7 nAChR signaling may offer novel strategies for combating influenza virus infections.


Asunto(s)
Pulmón , Infecciones por Orthomyxoviridae , Transducción de Señal , Nervio Vago , Receptor Nicotínico de Acetilcolina alfa 7 , Animales , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/genética , Nervio Vago/metabolismo , Ratones , Infecciones por Orthomyxoviridae/virología , Pulmón/virología , Pulmón/patología , Ratones Endogámicos C57BL , Células Epiteliales Alveolares/virología , Células Epiteliales Alveolares/metabolismo , Humanos , Ratones Noqueados
19.
Front Immunol ; 15: 1394690, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38994372

RESUMEN

Connective tissue represents the support matrix and the connection between tissues and organs. In its composition, collagen, the major structural protein, is the main component of the skin, bones, tendons and ligaments. Especially at the pediatric age, its damage in the context of pathologies such as systemic lupus erythematosus, scleroderma or dermatomyositis can have a significant negative impact on the development and optimal functioning of the body. The consequences can extend to various structures (e.g., joints, skin, eyes, lungs, heart, kidneys). Of these, we retain and reveal later in our manuscript, mainly the respiratory involvement. Manifested in various forms that can damage the chest wall, pleura, interstitium or vascularization, lung damage in pediatric systemic inflammatory diseases is underdeveloped in the literature compared to that described in adults. Under the threat of severe evolution, sometimes rapidly progressive and leading to death, it is necessary to increase the popularization of information aimed at physiopathological triggering and maintenance mechanisms, diagnostic means, and therapeutic directions among medical specialists. In addition, we emphasize the need for interdisciplinary collaboration, especially between pediatricians, rheumatologists, infectious disease specialists, pulmonologists, and immunologists. Through our narrative review we aimed to bring up to date, in a concise and easy to assimilate, general principles regarding the pulmonary impact of collagenoses using the most recent articles published in international libraries, duplicated by previous articles, of reference for the targeted pathologies.


Asunto(s)
Enfermedades del Colágeno , Humanos , Niño , Enfermedades del Colágeno/complicaciones , Pulmón/patología , Pulmón/inmunología , Enfermedades Pulmonares/etiología , Morbilidad
20.
Clin Respir J ; 18(7): e13807, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38994638

RESUMEN

The gradually progressive solitary cystic-solid mass of chest CT scans is highly suggestive of lung cancer. We report a case of a 29-year-old woman with a persistent cystic-solid lesion in the right upper lobe. A chest CT scan showed a 35 mm × 44 mm × 51 mm focal cystic-solid mass in the anterior segment of the right upper lobe. The size of lesion had increased over 3 years, especially for the solid component. The right upper lobe pneumonectomy was performed. Postoperative pathological examination showed placental transmogrification of the lung, which is a rare cause of pulmonary cystic lesion.


Asunto(s)
Neumonectomía , Tomografía Computarizada por Rayos X , Humanos , Femenino , Adulto , Tomografía Computarizada por Rayos X/métodos , Neumonectomía/métodos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/cirugía , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/diagnóstico , Pulmón/diagnóstico por imagen , Pulmón/patología , Pulmón/cirugía , Diagnóstico Diferencial , Embarazo , Enfermedades Pulmonares/cirugía , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/diagnóstico por imagen , Enfermedades Pulmonares/diagnóstico , Quistes/cirugía , Quistes/patología , Quistes/diagnóstico por imagen , Quistes/diagnóstico , Coristoma/cirugía , Coristoma/patología , Coristoma/diagnóstico , Coristoma/diagnóstico por imagen , Resultado del Tratamiento , Placenta/patología , Placenta/diagnóstico por imagen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...