Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.037
Filtrar
1.
J Ethnopharmacol ; 336: 118654, 2025 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-39098621

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Chaihu Guizhi Decoction (CGD) has a long history of use in China for the treatment of influenza, which involves the use of a variety of aromatic herbs. Our previous studies have found that the contents of aromatic constituents in CGD affected the efficacy of treatment of influenza-infected mice, suggesting a clue that essential oil from CGD may play a relatively important role in ameliorating influenza induced pneumonia. AIM OF THE STUDY: To evaluate the anti-influenza potential of essential oil derived from Chaihu Guizhi Decoction (CGD-EO), to characterize and predict the key active components in CGD-EO, and to explore the mechanism of action of CGD-EO. MATERIALS AND METHODS: CGD-EO was obtained by steam distillation, and the components of the essential oil were characterized by gas chromatography-mass spectrometry (GC-MS) in conjunction with the retention index. The constituents absorbed into the blood of mice treated with CGD-EO were analyzed by headspace solid phase microextraction gas chromatography/mass spectrometry (HS-SPME-GC/MS). The potential anti-influenza active constituents and their possible action pathway were predicted by simulation using a network pharmacology approach. The protective effect of CGD-EO and its major components on H1N1/PR8-infected cells was determined using the CCK8 assay kit. Mice infected with influenza A virus H1N1/PR8 were administered different doses of CGD-EO orally and the body weights and lung weights were recorded. Mice with varying degrees of H1N1/PR8 infection were administered CGD-EO orally, and their daily weight, water consumption, and clinical indicators were recorded. Necropsies were conducted on days 3 and 5, during which lung weights were measured and lung tissues were preserved. Furthermore, the mRNA expression of the H1N1/PR8 virus and inflammatory factors in lung tissue was analyzed using RT-qPCR. RESULTS: (E)-cinnamaldehyde was the most abundant compound in the CGD-EO. The results of serum medicinal chemistry combined with network pharmacological analysis indicated that (E)-cinnamaldehyde and 3-phenyl-2-propenal may be potential active components of the CGD-EO anti-influenza, and may be involved in the NF-κB signalling pathway. In vitro studies have demonstrated that both CGD-EO and cinnamaldehyde exert a protective effect on MDCK cells infected with H1N1/PR8. In a 0.5 TCID50 H1N1/PR8-induced influenza model, mice treated with CGD-EO at a dose of 63.50 µg/kg exhibited a reduction in lung index, pathological lung lesions, and H1N1/PR8 viral gene levels. In addition, CGD-EO treatment was found to regulate the levels of inflammatory cytokines, including IL-6, TNF-α, and IFN-γ. Moreover, following three days of administration, an upregulation of NF-κB mRNA levels in mouse lung tissue was observed in response to CGD-EO treatment. CONCLUSIONS: The findings of our study indicate CGD-EO exerts a protective effect against H1N1-induced cytopathic lesions in vitro and is capable of alleviating H1N1-induced pneumonitis in mice. Moreover, it appears to be more efficacious in the treatment of mild symptoms of H1N1 infection. Studies have demonstrated that CGD-EO has antiviral potential to attenuate influenza-induced lung injury by modulating inflammatory cytokines and NF-κB signalling pathways during the early stages of influenza infection. It is possible that (E)-cinnamaldehyde is a potential active ingredient in the anti-influenza efficacy of CGD-EO.


Assuntos
Antivirais , Medicamentos de Ervas Chinesas , Óleos Voláteis , Infecções por Orthomyxoviridae , Animais , Óleos Voláteis/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Camundongos , Infecções por Orthomyxoviridae/tratamento farmacológico , Antivirais/farmacologia , Camundongos Endogâmicos BALB C , Pneumonia Viral/tratamento farmacológico , Masculino , Células Madin Darby de Rim Canino , Cães , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/virologia , Pulmão/metabolismo , Humanos , Feminino , Pneumonia/tratamento farmacológico , Pneumonia/virologia , Pneumonia/metabolismo
2.
Front Immunol ; 15: 1432334, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39351225

RESUMO

Background: Environmental lipopolysaccharide (LPS) and microbial component-enriched organic dusts cause significant lung disease. These environmental exposures induce the recruitment and activation of distinct lung monocyte/macrophage subpopulations involved in disease pathogenesis. Aconitate decarboxylase 1 (Acod1) was one of the most upregulated genes following LPS (vs. saline) exposure of murine whole lungs with transcriptomic profiling of sorted lung monocyte/macrophage subpopulations also highlighting its significance. Given monocyte/macrophage activation can be tightly linked to metabolism, the objective of these studies was to determine the role of the immunometabolic regulator ACOD1 in environmental exposure-induced lung inflammation. Methods: Wild-type (WT) mice were intratracheally (i.t.) instilled with 10 µg of LPS or saline. Whole lungs were profiled using bulk RNA sequencing or sorted to isolate monocyte/macrophage subpopulations. Sorted subpopulations were then characterized transcriptomically using a NanoString innate immunity multiplex array 48 h post-exposure. Next, WT and Acod1-/- mice were instilled with LPS, 25% organic dust extract (ODE), or saline, whereupon serum, bronchoalveolar lavage fluid (BALF), and lung tissues were collected. BALF metabolites of the tricarboxylic acid (TCA) cycle were quantified by mass spectrometry. Cytokines/chemokines and tissue remodeling mediators were quantitated by ELISA. Lung immune cells were characterized by flow cytometry. Invasive lung function testing was performed 3 h post-LPS with WT and Acod1-/- mice. Results: Acod1-/- mice treated with LPS demonstrated decreased BALF levels of itaconate, TCA cycle reprogramming, decreased BALF neutrophils, increased lung CD4+ T cells, decreased BALF and lung levels of TNF-α, and decreased BALF CXCL1 compared to WT animals. In comparison, Acod1-/- mice treated with ODE demonstrated decreased serum pentraxin-2, BALF levels of itaconate, lung total cell, neutrophil, monocyte, and B-cell infiltrates with decreased BALF levels of TNF-α and IL-6 and decreased lung CXCL1 vs. WT animals. Mediators of tissue remodeling (TIMP1, MMP-8, MMP-9) were also decreased in the LPS-exposed Acod1-/- mice, with MMP-9 also reduced in ODE-exposed Acod1-/- mice. Lung function assessments demonstrated a blunted response to LPS-induced airway hyperresponsiveness in Acod1-/- animals. Conclusion: Acod1 is robustly upregulated in the lungs following LPS exposure and encodes a key immunometabolic regulator. ACOD1 mediates the proinflammatory response to acute inhaled environmental LPS and organic dust exposure-induced lung inflammation.


Assuntos
Carboxiliases , Lipopolissacarídeos , Camundongos Knockout , Animais , Camundongos , Carboxiliases/metabolismo , Carboxiliases/genética , Lipopolissacarídeos/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Camundongos Endogâmicos C57BL , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Exposição Ambiental/efeitos adversos , Pneumonia/imunologia , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Citocinas/metabolismo , Masculino , Hidroliases
3.
FASEB J ; 38(18): e70051, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39269436

RESUMO

Pseudomonas aeruginosa is a frequent cause of antimicrobial-resistant hospital-acquired pneumonia, especially in critically ill patients. Inflammation triggered by P. aeruginosa infection is necessary for bacterial clearance but must be spatially and temporally regulated to prevent further tissue damage and bacterial dissemination. Emerging data have shed light on the pro-resolving actions of angiotensin-(1-7) [Ang-(1-7)] signaling through the G protein-coupled receptor Mas (MasR) during infections. Herein, we investigated the role of the Ang-(1-7)/Mas axis in pneumonia caused by P. aeruginosa by using genetic and pharmacological approach and found that Mas receptor-deficient animals developed a more severe form of pneumonia showing higher neutrophilic infiltration into the airways, bacterial load, cytokines, and chemokines production and more severe pulmonary damage. Conversely, treatment of pseudomonas-infected mice with Ang-(1-7) was able to decrease neutrophilic infiltration in airways and lungs, local and systemic levels of pro-inflammatory cytokines and chemokines, and increase the efferocytosis rates, mitigating lung damage/dysfunction caused by infection. Notably, the therapeutic association of Ang-(1-7) with antibiotics improved the survival rates of mice subjected to lethal inoculum of P. aeruginosa, extending the therapeutic window for imipenem. Mechanistically, Ang-(1-7) increased phagocytosis of bacteria by neutrophils and macrophages to accelerate pathogen clearance. Altogether, harnessing the Ang-(1-7) pathway during infection is a potential strategy for the development of host-directed therapies to promote mechanisms of resistance and resilience to pneumonia.


Assuntos
Angiotensina I , Antibacterianos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos , Proto-Oncogene Mas , Infecções por Pseudomonas , Pseudomonas aeruginosa , Receptores Acoplados a Proteínas G , Animais , Angiotensina I/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos , Camundongos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/microbiologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Pneumonia Bacteriana/tratamento farmacológico , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Pneumonia Bacteriana/metabolismo , Citocinas/metabolismo , Camundongos Knockout , Pneumonia/tratamento farmacológico , Pneumonia/metabolismo , Pneumonia/microbiologia , Masculino , Pulmão/microbiologia , Pulmão/metabolismo , Pulmão/patologia , Transdução de Sinais/efeitos dos fármacos , Infiltração de Neutrófilos/efeitos dos fármacos
4.
Commun Biol ; 7(1): 1090, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39237613

RESUMO

T cell immunoglobulin and mucin-containing molecule 3 (TIM-3) exhibits unique, cell type- and context-dependent characteristics and functions. Here, we report that TIM-3 on myeloid cells plays essential roles in modulating lung inflammation. We found that myeloid cell-specific TIM-3 knock-in (FSF-TIM3/LysM-Cre+) mice have lower body weight and shorter lifespan than WT mice. Intriguingly, the lungs of FSF-TIM3/LysM-Cre+ mice display excessive inflammation and features of disease-associated pathology. We further revealed that galectin-3 levels are notably elevated in TIM-3-overexpressing lung-derived myeloid cells. Furthermore, both TIM-3 blockade and GB1107, a galectin-3 inhibitor, ameliorated lung inflammation in FSF-TIM3/LysM-Cre+/- mice. Using an LPS-induced lung inflammation model with myeloid cell-specific TIM-3 knock-out mice, we demonstrated the association of TIM-3 with both lung inflammation and galectin-3. Collectively, our findings suggest that myeloid TIM-3 is an important regulator in the lungs and that modulation of TIM-3 and galectin-3 could offer therapeutic benefits for inflammation-associated lung diseases.


Assuntos
Galectina 3 , Receptor Celular 2 do Vírus da Hepatite A , Células Mieloides , Pneumonia , Animais , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/genética , Galectina 3/metabolismo , Galectina 3/genética , Células Mieloides/metabolismo , Camundongos , Pneumonia/metabolismo , Pneumonia/patologia , Pneumonia/genética , Camundongos Knockout , Camundongos Endogâmicos C57BL , Galectinas/metabolismo , Galectinas/genética , Pulmão/patologia , Pulmão/metabolismo
5.
Cells ; 13(18)2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39329762

RESUMO

BACKGROUND: Despite promise in preclinical models of acute respiratory distress syndrome (ARDS), mesenchymal stem cells (MSC) have failed to translate to therapeutic benefit in clinical trials. The MSC is a live cell medicine and interacts with the patient's disease state. Here, we explored this interaction, seeking to devise strategies to enhance MSC therapeutic function. METHODS: Human bone-marrow-derived MSCs were exposed to lung homogenate from healthy and E. coli-induced ARDS rat models. Apoptosis and functional assays of the MSCs were performed. RESULTS: The ARDS model showed reduced arterial oxygenation, decreased lung compliance and an inflammatory microenvironment compared to controls. MSCs underwent more apoptosis after stimulation by lung homogenate from controls compared to E. coli, which may explain why MSCs persist longer in ARDS subjects after administration. Changes in expression of cell surface markers and cytokines were associated with lung homogenate from different groups. The anti-microbial effects of MSCs did not change with the stimulation. Moreover, the conditioned media from lung-homogenate-stimulated MSCs inhibited T-cell proliferation. CONCLUSIONS: These findings suggest that the ARDS microenvironment plays an important role in the MSC's therapeutic mechanism of action, and changes can inform strategies to modulate MSC-based cell therapy for ARDS.


Assuntos
Microambiente Celular , Pulmão , Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Humanos , Pulmão/patologia , Ratos , Síndrome do Desconforto Respiratório/terapia , Síndrome do Desconforto Respiratório/patologia , Síndrome do Desconforto Respiratório/metabolismo , Pneumonia/patologia , Pneumonia/metabolismo , Pneumonia/terapia , Masculino , Apoptose , Proliferação de Células , Ratos Sprague-Dawley , Modelos Animais de Doenças , Escherichia coli , Citocinas/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Linfócitos T/imunologia , Linfócitos T/metabolismo
6.
Cells ; 13(17)2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39272979

RESUMO

Circular RNA (circRNA) is a type of single-stranded RNA that forms a covalently closed continuous loop, unlike linear RNA. The expression of circRNAs in mammals is often conserved across species and shows tissue and cell specificity. Some circRNA serve as gene regulators. However, the biological function of most circRNAs is unclear. CircRNA does not have 5' or 3' ends. The unique structure of circRNAs provides them with a much longer half-life and more resistance to RNase R than linear RNAs. Inflammatory lung responses occur in the pathogenesis and recovery of many lung diseases. Macrophages form the first line of host defense/innate immune responses and initiate/mediate lung inflammation. For example, in bacterial pneumonia, upon pro-inflammatory activation, they release early response cytokines/chemokines that recruit neutrophils, macrophages, and lymphocytes to sites of infection and clear pathogens. The functional effects and mechanisms by which circRNAs exert physiological or pathological roles in macrophage activation and lung inflammation remain poorly understood. In this article, we will review the current understanding and progress of circRNA biogenesis, regulation, secretion, and degradation. Furthermore, we will review the current reports on the role of circRNAs in macrophage activation and polarization, as well as in the process of inflammatory lung responses.


Assuntos
Pulmão , Ativação de Macrófagos , RNA Circular , RNA Circular/genética , RNA Circular/metabolismo , Humanos , Ativação de Macrófagos/genética , Animais , Pulmão/patologia , Pulmão/metabolismo , Pulmão/imunologia , Macrófagos/metabolismo , Macrófagos/imunologia , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , Inflamação/genética , Inflamação/patologia
7.
J Innate Immun ; 16(1): 397-412, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39134014

RESUMO

INTRODUCTION: MDM2 is known as the primary negative regulator of p53, and MDM2 promotes lung cancer fibrosis and lung injury through p53-dependent and p53-independent pathways. However, the mechanism by which MDM2 influences the pathogenesis of asthma is unknown. In this study, we investigated the function of MDM2 in lung epithelial cells in type 2 lung inflammation. METHODS: We used type II alveolar epithelial cell-specific heterozygous knockout of Mdm2 mice to validate its function. Then papain-induced asthma model was established, and changes in inflammation were observed by measuring immunohistochemistry and flow cytometry analysis. RESULTS: In this study, we knockdown the mouse Mdm2 gene in type 2 alveolar epithelial cells. We demonstrated that heterozygous Mdm2 gene-deleted mice were highly susceptible to protease allergen papain-induced pulmonary inflammation characterized by increased ILC2 numbers, IL-5 and IL-13 cytokine levels, and lung pathology. A mechanistic study showed that following the decreased expression of Mdm2 in lung epithelial cells and A549 cell line, p53 was overactivated, and the expression of its downstream genes p21, Puma, and Noxa was elevated, which resulted in apoptosis. After Mdm2 knockdown, the mRNA expression of inflammation-related gene IL-25, HMGB1, and TNF-α were increased, which further amplified the downstream ILC2 response and lung inflammation. CONCLUSION: These results indicate that Mdm2 maintains the homeostasis of lung epithelial cells by targeting P53 and regulates the function of lung epithelial cells under type 2 lung inflammation.


Assuntos
Asma , Homeostase , Camundongos Knockout , Proteínas Proto-Oncogênicas c-mdm2 , Proteína Supressora de Tumor p53 , Animais , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Camundongos , Humanos , Asma/imunologia , Asma/metabolismo , Asma/induzido quimicamente , Asma/genética , Células A549 , Modelos Animais de Doenças , Apoptose , Células Epiteliais/metabolismo , Células Epiteliais Alveolares/metabolismo , Papaína , Camundongos Endogâmicos C57BL , Pneumonia/imunologia , Pneumonia/metabolismo
8.
Immunology ; 173(3): 590-602, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39132816

RESUMO

Interleukin (IL)-33 released from airway epithelial cells plays a vital role in shaping type 2 immune responses by binding to the ST2 receptor present in many immune cells, including mast cells (MCs). Intranasal administration of IL-33 in mice induces type 2 lung inflammation, an increase in lung MC progenitors, and transepithelial migration of leukocytes to the bronchoalveolar space. The aim of this study was to determine the contribution of MCs in IL-33-induced lung pathology. Four daily intranasal administrations of IL-33 reduced spirometry-like lung function parameters, induced airway hyperresponsiveness, and increased leukocytes in bronchoalveolar lavage fluid (BAL) in an ST2-dependent manner. MC-deficient (Cpa3cre/+) mice, which lack MCs, had intact spirometry-like lung function but slightly reduced airway hyperresponsiveness, possibly related to reduced IL-33 or serotonin. Strikingly, Cpa3cre/+ mice exposed to IL-33 had 50% reduction in BAL T-cells, and CXCL1 and IL-33 were reduced in the lung. Intranasal IL-33 induced CXCR2 expression in T-cells in a MC-independent fashion. Furthermore, IL-33-induced lung MCs were immunopositive for CXCL1 and localized in the epithelium of wild-type mice. These results suggest that MCs are required to sustain intact lung IL-33 and CXCL1 levels in mice with IL-33-induced airway inflammation, thereby facilitating T-cell accumulation in the bronchoalveolar space.


Assuntos
Líquido da Lavagem Broncoalveolar , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Mastócitos , Camundongos Knockout , Linfócitos T , Animais , Interleucina-33/metabolismo , Interleucina-33/imunologia , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Camundongos Endogâmicos C57BL , Pulmão/imunologia , Pulmão/patologia , Pneumonia/imunologia , Pneumonia/metabolismo , Receptores de Interleucina-8B/metabolismo , Quimiocina CXCL1/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 44(10): 2213-2222, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-39145395

RESUMO

BACKGROUND: Platelets prevent bleeding in a variety of inflammatory settings, the adhesion receptors and activation pathways involved being highly context-dependent and functionally redundant. In some situations, platelets recruited to inflammatory sites act independently of aggregation. The mechanisms underlying stable platelet adhesion in inflamed microvessels remain incompletely understood, in particular, whether and if so, how ß1 and ß3 integrins are involved. METHODS: The impact of isolated or combined platelet deficiency in ß1 and ß3 integrins on inflammation-associated hemostasis was investigated in 3 models of acute inflammation: immune complex-based cutaneous reverse passive Arthus reaction, intranasal lipopolysaccharide-induced lung inflammation, and cerebral ischemia-reperfusion following transient (2-hour) occlusion of the middle cerebral artery. RESULTS: Mice with platelet-directed inactivation of Itgb1 (PF4Cre-ß1-/-) displayed no bleeding in any of the inflammation models, while mice defective in platelet Itgb3 (PF4Cre-ß3-/-) exhibited bleeding in all 3 models. Remarkably, the bleeding phenotype of PF4Cre-ß3-/- mice was exacerbated in the reverse passive Arthus model by the concomitant deletion of ß1 integrins, PF4Cre-ß1-/-/ß3-/- animals presenting increased bleeding. Intravital microscopy in reverse passive Arthus experiments highlighted a major defect in the adhesion of PF4Cre-ß1-/-/ß3-/- platelets to inflamed microvessels. Unlike PF4Cre-ß1-/- and PF4Cre-ß3-/- mice, PF4Cre-ß1-/-/ß3-/- animals developed early hemorrhagic transformation 6 hours after transient middle cerebral artery occlusion. PF4Cre-ß1-/-/ß3-/- mice displayed no more bleeding in lipopolysaccharide-induced lung inflammation than PF4Cre-ß3-/- animals. CONCLUSIONS: Altogether, these results show that the requirement for and degree of functional redundancy between platelet ß1 and ß3 integrins in inflammation-associated hemostasis vary with the inflammatory situation.


Assuntos
Plaquetas , Modelos Animais de Doenças , Hemorragia , Integrina beta1 , Integrina beta3 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Animais , Masculino , Camundongos , Plaquetas/metabolismo , Hemorragia/genética , Hemorragia/sangue , Hemostasia , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/sangue , Infarto da Artéria Cerebral Média/metabolismo , Inflamação/genética , Inflamação/metabolismo , Inflamação/sangue , Integrina beta1/metabolismo , Integrina beta1/genética , Integrina beta3/genética , Integrina beta3/metabolismo , Lipopolissacarídeos , Adesividade Plaquetária , Pneumonia/genética , Pneumonia/sangue , Pneumonia/metabolismo , Pneumonia/patologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/sangue
10.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167472, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39154794

RESUMO

COVID-19, caused by SARS-CoV-2 infection, results in irreversible or fatal lung injury. We assumed that necroptosis of virus-infected alveolar epithelial cells (AEC) could promote local inflammation and further lung injury in COVID-19. Since CD8+ lymphocytes induced AEC cell death via cytotoxic molecules such as FAS ligands, we examined the involvement of FAS-mediated cell death in COVID-19 patients and murine COVID-19 model. We identified the occurrence of necroptosis and subsequent release of HMGB1 in the admitted patients with COVID-19. In the mouse model of COVID-19, lung inflammation and injury were attenuated in Fas-deficient mice compared to Fas-intact mice. The infection enhanced Type I interferon-inducible genes in both groups, while inflammasome-associated genes were specifically upregulated in Fas-intact mice. The treatment with necroptosis inhibitor, Nec1s, improved survival rate, lung injury, and systemic inflammation. SARS-CoV-2 induced necroptosis causes cytokine induction and lung damage, and its inhibition could be a novel therapeutic strategy for COVID-19.


Assuntos
Células Epiteliais Alveolares , COVID-19 , Necroptose , SARS-CoV-2 , COVID-19/patologia , COVID-19/imunologia , COVID-19/metabolismo , COVID-19/virologia , COVID-19/complicações , Animais , Humanos , Camundongos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Células Epiteliais Alveolares/virologia , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Lesão Pulmonar/patologia , Lesão Pulmonar/virologia , Lesão Pulmonar/imunologia , Lesão Pulmonar/metabolismo , Masculino , Modelos Animais de Doenças , Feminino , Camundongos Endogâmicos C57BL , Receptor fas/metabolismo , Receptor fas/genética , Camundongos Knockout , Pneumonia/patologia , Pneumonia/virologia , Pneumonia/metabolismo , Pneumonia/imunologia , Pessoa de Meia-Idade , Imidazóis , Indóis
11.
Nat Commun ; 15(1): 7396, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39191805

RESUMO

Many lung immune cells are known to respond to inhaled particulate matter. However, current known responses cannot explain how particles induce thrombosis in the lung and how they translocate to distant organs. Here, we demonstrate that lung megakaryocytes (MKs) in the alveolar and interstitial regions display location-determined characteristics and act as crucial responders to inhaled particles. They move rapidly to engulf particles and become activated with upregulation in inflammatory responses and thrombopoiesis. Comprehensive in vivo, in vitro and ex vivo results unraveled that MKs were involved in particle-induced lung damages and shed particle-containing platelets into blood circulation. Moreover, MK-derived platelets exhibited faster clotting, stronger adhesion than normal resting platelets, and inherited the engulfed particles from parent MKs to assist in extrapulmonary particle transportation. Our findings collectively highlight that the specific responses of MKs towards inhaled particles and their roles in facilitating the translocation of particles from the lungs to extrapulmonary organs for clearance.


Assuntos
Plaquetas , Pulmão , Megacariócitos , Camundongos Endogâmicos C57BL , Material Particulado , Animais , Pulmão/patologia , Pulmão/imunologia , Plaquetas/metabolismo , Camundongos , Masculino , Pneumonia/patologia , Pneumonia/imunologia , Pneumonia/metabolismo , Trombopoese , Humanos
12.
Nat Commun ; 15(1): 7241, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39174557

RESUMO

Type 2 alveolar epithelial (AT2) cells of the lung are fundamental in regulating alveolar inflammation in response to injury. Impaired mitochondrial long-chain fatty acid ß-oxidation (mtLCFAO) in AT2 cells is assumed to aggravate alveolar inflammation in acute lung injury (ALI), yet the importance of mtLCFAO to AT2 cell function needs to be defined. Here we show that expression of carnitine palmitoyltransferase 1a (CPT1a), a mtLCFAO rate limiting enzyme, in AT2 cells is significantly decreased in acute respiratory distress syndrome (ARDS). In mice, Cpt1a deletion in AT2 cells impairs mtLCFAO without reducing ATP production and alters surfactant phospholipid abundance in the alveoli. Impairing mtLCFAO in AT2 cells via deleting either Cpt1a or Acadl (acyl-CoA dehydrogenase long chain) restricts alveolar inflammation in ALI by hindering the production of the neutrophilic chemokine CXCL2 from AT2 cells. This study thus highlights mtLCFAO as immunometabolism to injury in AT2 cells and suggests impaired mtLCFAO in AT2 cells as an anti-inflammatory response in ARDS.


Assuntos
Lesão Pulmonar Aguda , Células Epiteliais Alveolares , Carnitina O-Palmitoiltransferase , Ácidos Graxos , Mitocôndrias , Oxirredução , Síndrome do Desconforto Respiratório , Animais , Carnitina O-Palmitoiltransferase/metabolismo , Carnitina O-Palmitoiltransferase/genética , Mitocôndrias/metabolismo , Células Epiteliais Alveolares/metabolismo , Ácidos Graxos/metabolismo , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/genética , Camundongos , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/patologia , Síndrome do Desconforto Respiratório/genética , Masculino , Humanos , Quimiocina CXCL2/metabolismo , Quimiocina CXCL2/genética , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Neutrófilos/metabolismo , Camundongos Knockout , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Acil-CoA Desidrogenase de Cadeia Longa/genética , Inflamação/metabolismo , Inflamação/patologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Alvéolos Pulmonares/imunologia , Trifosfato de Adenosina/metabolismo , Pneumonia/metabolismo , Pneumonia/imunologia , Pneumonia/patologia , Pneumonia/genética
13.
Biomed Pharmacother ; 178: 116992, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39106709

RESUMO

The effective treatment of acute lung injury (ALI) remains a significant challenge. Patients with ALI demonstrate an abundance of proinflammatory mediators in both bronchoalveolar lavage fluid (BALF) and circulating plasma. Bardoxolone methyl (BM) is a semi-synthetic triterpenoid derived from oleanolic acid, a natural product known for its ability to inhibit proinflammatory signaling. GSDMD is a signaling protein involved in pyroptosis, a form of programmed cell death. It has been reported that its upstream proteins play a role in the pathogenesis of ALI. However, there is currently no research examining whether the effect of BM on the occurrence and development of ALI is associated with changes in GSDMD protein. In this study, we prepared nanostructured lipid carriers loaded with BM and conjugated with anti-PECAM-1 antibody (PECAM@BM NLCs). PECAM@BM NLCs were designed to specifically bind to pulmonary vascular endothelial cells that highly express the PECAM-1 receptors. We also aimed to investigate the protective effects of PECAM@BM NLCs on ALI and elucidate the underlying molecular mechanisms. The results demonstrated that PECAM@BM NLCs accumulated in the lung tissues and significantly alleviated the inflammatory injury of ALI. This was evidenced by the changes in the lung wet/dry ratio, the total protein concentration, proinflammatory cytokines in BALF, and the histopathological progress. Additionally, we elucidated that PECAM@BM NLCs had the ability to inhibit the assembly of NLRP3 inflammasome and pro-caspase-1 complex, thereby suppressing the induction of pyroptosis. This mechanism resulted in the inhibition of N-terminal GSDMD expression and effectively prevented the progression of ALI.


Assuntos
Lesão Pulmonar Aguda , Pulmão , Nanoestruturas , Ácido Oleanólico , Molécula-1 de Adesão Celular Endotelial a Plaquetas , Ácido Oleanólico/farmacologia , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/administração & dosagem , Ácido Oleanólico/química , Animais , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/patologia , Nanoestruturas/química , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Portadores de Fármacos/química , Masculino , Camundongos , Pneumonia/tratamento farmacológico , Pneumonia/patologia , Pneumonia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Camundongos Endogâmicos C57BL , Lipídeos/química , Anticorpos/farmacologia , Líquido da Lavagem Broncoalveolar/química , Humanos , Sistemas de Liberação de Medicamentos/métodos , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos
14.
Immunobiology ; 229(5): 152843, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39186867

RESUMO

We previously reported that myeloperoxidase-deficient (MPO-/-) mice develop more severe neutrophil-rich lung inflammation than wild-type mice following intranasal Zymosan administration. Interestingly, we found that these mutant mice with severe lung inflammation also displayed pronounced neutrophilia and anemia, characterized by increased granulopoiesis and decreased erythropoiesis in the bone marrow, compared to wild-type mice. This condition was associated with higher concentrations of granulocyte-colony stimulating factor (G-CSF) in both the lungs and serum, a factor known to enhance granulopoiesis. Neutrophils accumulating in the lungs of MPO-/- mice produced greater amounts of G-CSF than those in wild-type mice, indicating that they are a significant source of G-CSF. In vitro experiments using signal transduction inhibitors and Western blot analysis revealed that MPO-/- neutrophils express higher levels of G-CSF mRNA in response to Zymosan, attributed to the upregulation of the IκB kinase/nuclear factor (NF)-κB pathway and the extracellular-signal-regulated kinase/NF-κB pathway. These findings highlight MPO as a critical regulator of granulopoiesis and erythropoiesis in inflamed tissues.


Assuntos
Anemia , Eritropoese , Fator Estimulador de Colônias de Granulócitos , Camundongos Knockout , Neutrófilos , Peroxidase , Pneumonia , Zimosan , Animais , Camundongos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Peroxidase/metabolismo , Anemia/etiologia , Pneumonia/etiologia , Pneumonia/metabolismo , Pneumonia/imunologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Transdução de Sinais , NF-kappa B/metabolismo , Granulócitos/metabolismo , Granulócitos/imunologia , Pulmão/patologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
15.
EBioMedicine ; 106: 105267, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39098109

RESUMO

BACKGROUND: Checkpoint inhibitor pneumonitis (CIP) is a potentially fatal adverse event characterized by new pulmonary infiltrates in cancer patients receiving immune checkpoint inhibitor therapy. This study aims to explore the interplay between lung microbiota, dysregulated metabolites, and host immunity in CIP. METHODS: We recruited thirteen hospitalized CIP patients, eleven idiopathic pulmonary fibrosis (IPF) patients, and ten new-onset non-small cell lung cancer patients. Bronchoalveolar lavage fluid samples were collected for 16S rRNA gene sequencing. The percentages of immune cells were determined using manual counting and flow cytometry. Interactions among microbiota, metabolites, and lymphocytes were analyzed using cultured mouse splenocytes and human T cells. FINDINGS: Proteobacteria emerged as the dominant phylum, notably abundant in both the CIP and IPF groups. Vibrio, Halomonas, Mangrovibacter, and Salinivibrio were the predominant microbiota because of their discriminative abundance patterns. Vibrio (r = 0.72, P-adj = 0.007) and Halomonas (r = 0.65, P-adj = 0.023) demonstrated strong correlations with lymphocytes. Vibrio metschnikovii and Mangrovibacter plantisponsors were more abundant in the CIP group than in the IPF group. Lauroylcarnitine, a key intermediary metabolite co-occurring with the predominant microbiota, exhibited a potent effect on cytokine secretion by mouse and human T cells, notably enhancing IFN-γ and TNF-α production from CD4 and CD8 cells in vitro. INTERPRETATION: Lauroylcarnitine, co-occurring with the predominant lung microbiota in CIP, could activate T cells in vitro. These findings suggest potential involvement of lung microbiota and acylcarnitine metabolism dysregulation in the pathogenesis of CIP. FUNDING: This work was supported by Peking University People's Hospital Scientific Research Development Funds (RDJ2022-15) and Provincial Key Clinical Specialty Capacity Building Project 2020 (Department of the Respiratory Medicine).


Assuntos
Inibidores de Checkpoint Imunológico , Pulmão , Ativação Linfocitária , Microbiota , Pneumonia , Linfócitos T , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Animais , Camundongos , Microbiota/efeitos dos fármacos , Masculino , Feminino , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Idoso , Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/patologia , Pulmão/imunologia , Pulmão/metabolismo , Pneumonia/microbiologia , Pneumonia/etiologia , Pneumonia/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pessoa de Meia-Idade , Carnitina/análogos & derivados , Carnitina/metabolismo , RNA Ribossômico 16S/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/microbiologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Citocinas/metabolismo
16.
Placenta ; 154: 220-223, 2024 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-39106563

RESUMO

BACKGROUND: Gestational diabetes mellitus (GDM) is a known risk factor for stillbirth (Rosenstein et al., 2012) [1]. Delayed villous maturation (DVM), predominantly seen in term placentas in pregnancies complicated with glucose dysmetabolism, may in part be a consequence of excessive maternal glucose leading to release of fetal insulin and other growth factors that promote excessive placental growth at the expense of villous maturation (Redline, 2012) [2]. CASES: We present three cases of under-diagnosed/treated glucose dysmetabolism in women in their first pregnancies cared for in other hospitals in the United Kingdom (UK) with the fatal fetal/neonatal outcomes and confirmed DVM in the placenta and congenital pneumonia on post-mortem examination in all three cases. CONCLUSION: This cluster supports a hypothesis that DVM and glucose dysmentabolism may make babies more susceptible to severe perinatal infection. All three cases received the antenatal care in their subsequent pregnancies in our unit and had confirmed glucose dysmetabolism which was treated and resulted in healthy babies.


Assuntos
Diabetes Gestacional , Pneumonia , Humanos , Feminino , Gravidez , Adulto , Pneumonia/metabolismo , Diabetes Gestacional/metabolismo , Recém-Nascido , Vilosidades Coriônicas/metabolismo , Vilosidades Coriônicas/patologia , Evolução Fatal , Doenças Fetais/metabolismo , Doenças Placentárias/metabolismo , Doenças Placentárias/patologia
17.
Food Chem Toxicol ; 191: 114893, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39067743

RESUMO

Particulate matter (PM) poses significant health risks, especially fine particles (PM2.5) that can cause severe lung injuries. Lupeol, a phytosterol from medicinal plants, has potential anti-cancer properties. This study investigated lupeol's protective effects against PM2.5-induced lung damage. Mice received lupeol following intratracheal PM2.5 exposure. Results showed lupeol reduced lung damage, lowered wet/dry (W/D) weight ratio, and suppressed increased permeability caused by PM2.5. Additionally, lupeol decreased plasma inflammatory cytokines, total protein concentration in bronchoalveolar lavage fluid (BALF), and PM2.5-induced lymphocyte proliferation. Lupeol also reduced expression of toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), and autophagy-related proteins microtubule-associated protein 1 A/1 B-light chain 3 (LC3) II and Beclin 1, while increasing phosphorylated mammalian target of rapamycin (mTOR) phosphorylation. These findings suggest lupeol's potential as a therapeutic agent for PM2.5-induced lung damage via modulation of the TLR4-MyD88 and mTOR-autophagy pathways.


Assuntos
Material Particulado , Triterpenos Pentacíclicos , Pneumonia , Receptor 4 Toll-Like , Animais , Triterpenos Pentacíclicos/farmacologia , Material Particulado/toxicidade , Receptor 4 Toll-Like/metabolismo , Pneumonia/tratamento farmacológico , Pneumonia/metabolismo , Pneumonia/induzido quimicamente , Camundongos , Masculino , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Serina-Treonina Quinases TOR/metabolismo , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Autofagia/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Citocinas/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Proteína Beclina-1/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Lupanos
18.
J Ethnopharmacol ; 335: 118609, 2024 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-39053707

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Xiebai San (XBS), a classic Chinese prescription, has been used for the clinical treatment of pneumonia-related diseases for thousands of years. However, the anti-pneumonia pharmacodynamic material basis of XBS and its underlying mechanisms remain unclear. AIM OF THE STUDY: This study aimed to comprehensively investigate and verify the anti-pneumonia pharmacodynamic material basis and mechanisms of XBS. MATERIALS AND METHODS: This study explored the anti-pneumonia activity and key pneumonia targets of XBS in lipopolysaccharide (LPS)-induced zebrafish and RAW264.7 cells in vivo and in vitro through transcriptomics, western blotting, and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The chemical fingerprint of XBS was established using high-performance liquid chromatography, and the similarities and areas of characteristic peaks of 15 batches of XBS were analyzed. Based on the spectrum-efficacy relationship, the potential anti-inflammatory components were screened according to their peak areas and efficacy using principal component analysis (PCA), bivariate correlation, and partial least squares regression analysis. Active components that bind to core targets were further screened based on surface plasmon resonance (SPR). The binding mode of proteins and components was simulated via molecular docking, which enabled the identification of the primary active components of XBS, thereby elucidating its anti-pneumonia properties. Finally, the anti-inflammatory activities of these components were verified in vitro. RESULTS: XBS decreased neutrophil aggregation in zebrafish and nitric oxide (NO) secretion in RAW264.7 cells as well as suppressed the release of downstream inflammatory cytokines such as iNOS, TNF-α, IL-1ß, IL-18, and CXCL10 related to TNF and JAK-STAT signaling pathways. The phosphorylation of IκBα, Akt, and Stat3 was alleviated after XBS in cells. The fingerprint similarities of 15 batches of XBS ranged from 0.381 to 0.994, with a large difference. A total of 15 characteristic peaks were identified, and the relative standard deviation of their peak areas ranged from 24.1% to 70.7%. The results of in vitro anti-inflammatory activities of 15 batches of XBS showed that all samples inhibited the expression levels of NO and nine inflammatory markers. The anti-inflammatory index of 15 batches of XBS was determined to be 0.69-0.96 based on transformation of the anti-inflammatory rate and composite index method via PCA. The spectrum-efficacy relationship model of 15 characteristic peak areas and the anti-inflammatory index showed that 7 main potential active components were related to the anti-inflammatory activity of XBS. Moreover, four components (mulberroside A, isoquercitrin, liquiritigenin, and glycyrrhizic acid) screened based on SPR had different affinities toward TNFR1, Akt1, and Stat3 proteins, and the binding modes were elucidated via molecular docking. Finally, in LPS-induced RAW264.7 cells, all four active components (at a concentration of 60 µM) significantly inhibited the expression levels of NO and inflammatory markers. CONCLUSIONS: Based on the comprehensive strategy of spectrum-efficacy relationship and SPR, mulberroside A, isoquercitrin, liquiritigenin, and glycyrrhizic acid were identified as the primary pharmacodynamic active components involved in the anti-pneumonia activity of XBS and were found to intervene in TNF and JAK-STAT signaling pathways.


Assuntos
Anti-Inflamatórios , Medicamentos de Ervas Chinesas , Pneumonia , Ressonância de Plasmônio de Superfície , Peixe-Zebra , Animais , Células RAW 264.7 , Camundongos , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/química , Anti-Inflamatórios/farmacologia , Pneumonia/tratamento farmacológico , Pneumonia/metabolismo , Simulação de Acoplamento Molecular , Lipopolissacarídeos , Óxido Nítrico/metabolismo
19.
Cell Mol Life Sci ; 81(1): 287, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38970705

RESUMO

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.


Assuntos
Homeostase , Receptores X do Fígado , Macrófagos Alveolares , Pneumonia , Surfactantes Pulmonares , Transdução de Sinais , Animais , Receptores X do Fígado/metabolismo , Receptores X do Fígado/genética , Surfactantes Pulmonares/metabolismo , Camundongos , Pneumonia/metabolismo , Pneumonia/patologia , Macrófagos Alveolares/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pulmão/metabolismo , Pulmão/patologia , Células Epiteliais Alveolares/metabolismo , Asma/metabolismo , Asma/patologia , Asma/genética , Colesterol/metabolismo , Metabolismo dos Lipídeos , Fagocitose
20.
Int J Mol Sci ; 25(13)2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-39000529

RESUMO

Despite significant efforts toward improving therapy for septic shock, mortality remains high. Applying veno-arterial (V-A) extracorporeal membrane oxygenation (ECMO) in this context remains controversial. Since the cannulation of the femoral artery for V-A ECMO return leads to lower body hyperoxia, this study investigated the impact of V-A ECMO therapy on the intestinal and hepatic microcirculation during septic shock in a rodent model. Thirty male Lewis rats were randomly assigned to receive V-A ECMO therapy with low (60 mL/kg/min) or high (90 mL/kg/min) blood flow or a sham procedure. Hemodynamic data were collected through a pressure-volume catheter in the left ventricle and a catheter in the lateral tail artery. Septic shock was induced by intravenous administration of lipopolysaccharide (1 mg/kg). The rats received lung-protective ventilation during V-A ECMO therapy. The hepatic and intestinal microcirculation was measured by micro-lightguide spectrophotometry after median laparotomy for two hours. Systemic and pulmonary inflammation was detected via enzyme-linked immunosorbent assays (ELISA) of the plasma and bronchoalveolar lavage (BAL), respectively, measuring tumor necrosis factor-alpha (TNF-α), interleukins 6 (IL-6) and 10 (IL-10), and C-X-C motif ligands 2 (CXCL2) and 5 (CXCL5). Oxygen saturation and relative hemoglobin concentration were reduced in the hepatic and intestinal microcirculation during V-A ECMO therapy, independent of the blood flow rate. Further, rats treated with V-A ECMO therapy also presented elevated systolic, diastolic, and mean arterial blood pressure and increased stroke volume, cardiac output, and left ventricular end-diastolic volume. However, left ventricular end-diastolic pressure was only elevated during high-flow V-A ECMO therapy. Blood gas analysis revealed a dilutional anemia during V-A ECMO therapy. ELISA analysis showed an elevated plasma CXCL2 concentration only during high-flow V-A ECMO therapy and elevated BAL CXCL2 and CXCL5 concentrations only during low-flow V-A ECMO therapy. Rats undergoing V-A ECMO therapy exhibited impaired microcirculation of the intestine and liver during septic shock despite increased blood pressure and cardiac output. Increased pulmonary inflammation was detected only during low-flow V-A ECMO therapy in septic shock.


Assuntos
Modelos Animais de Doenças , Oxigenação por Membrana Extracorpórea , Intestinos , Fígado , Microcirculação , Ratos Endogâmicos Lew , Choque Séptico , Animais , Oxigenação por Membrana Extracorpórea/métodos , Masculino , Ratos , Choque Séptico/terapia , Choque Séptico/fisiopatologia , Choque Séptico/metabolismo , Fígado/metabolismo , Fígado/irrigação sanguínea , Intestinos/irrigação sanguínea , Pneumonia/terapia , Pneumonia/metabolismo , Pneumonia/fisiopatologia , Hemodinâmica , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA