Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.845
Filtrar
1.
Nat Cardiovasc Res ; 3(9): 1158-1176, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39271818

RESUMEN

The innate immune response is triggered rapidly after injury and its spatiotemporal dynamics are critical for regeneration; however, many questions remain about its exact role. Here we show that MyD88, a key component of the innate immune response, controls not only the inflammatory but also the fibrotic response during zebrafish cardiac regeneration. We find in cryoinjured myd88-/- ventricles a significant reduction in neutrophil and macrophage numbers and the expansion of a collagen-rich endocardial population. Further analyses reveal compromised PI3K/AKT pathway activation in the myd88-/- endocardium and increased myofibroblasts and scarring. Notably, endothelial-specific overexpression of myd88 reverses these neutrophil, fibrotic and scarring phenotypes. Mechanistically, we identify the endocardial-derived chemokine gene cxcl18b as a target of the MyD88 signaling pathway, and using loss-of-function and gain-of-function tools, we show that it controls neutrophil recruitment. Altogether, these findings shed light on the pivotal role of MyD88 in modulating inflammation and fibrosis during tissue regeneration.


Asunto(s)
Fibrosis , Inmunidad Innata , Factor 88 de Diferenciación Mieloide , Regeneración , Transducción de Señal , Proteínas de Pez Cebra , Pez Cebra , Animales , Animales Modificados Genéticamente , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Endocardio/metabolismo , Endocardio/patología , Endocardio/inmunología , Corazón/fisiopatología , Inmunidad Innata/genética , Macrófagos/metabolismo , Macrófagos/inmunología , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patología , Infiltración Neutrófila , Neutrófilos/metabolismo , Neutrófilos/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regeneración/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
2.
FASEB J ; 38(18): e70051, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39269436

RESUMEN

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.


Asunto(s)
Angiotensina I , Antibacterianos , Ratones Endogámicos C57BL , Fragmentos de Péptidos , Proto-Oncogenes Mas , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Receptores Acoplados a Proteínas G , Animales , Angiotensina I/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Ratones , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Neumonía Bacteriana/metabolismo , Citocinas/metabolismo , Ratones Noqueados , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Neumonía/microbiología , Masculino , Pulmón/microbiología , Pulmón/metabolismo , Pulmón/patología , Transducción de Señal/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos
3.
Sci Rep ; 14(1): 19870, 2024 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-39191825

RESUMEN

Neutrophil infiltration plays a key role in the pathogenesis of chronic rhinosinusitis with nasal polyps (CRSwNP). However, pertinent mechanisms remain poorly elucidated. Here, we obtained the data from gene expression omnibus (GEO) and gene set enrichment analysis (GSEA) to identify and validate neutrophil-associated hub genes in CRSwNP. We found that four neutrophil-associated hub genes, namely ICAM1, IL-1ß, TYROBP, and BCL2A1, were markedly upregulated and positively correlated with neutrophil infiltration levels in patients with CRSwNP. Subsequently, this was confirmed by real-time quantitative PCR. In conclusion, we identified the role of neutrophil infiltration in the pathophysiology of CRSwNP, which may be the potential targets for the diagnosis and treatment of CRSwNP.


Asunto(s)
Pólipos Nasales , Neutrófilos , Rinitis , Sinusitis , Pólipos Nasales/genética , Humanos , Sinusitis/genética , Rinitis/genética , Neutrófilos/metabolismo , Enfermedad Crónica , Interleucina-1beta/genética , Molécula 1 de Adhesión Intercelular/genética , Infiltración Neutrófila/genética , Masculino , Femenino , Perfilación de la Expresión Génica , Rinosinusitis
4.
Int Immunopharmacol ; 140: 112888, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39133956

RESUMEN

Pancreatic cancer is characterized by an extremely poor prognosis and presents significant treatment challenges. Liver metastasis is the leading cause of death in patients with pancreatic cancer. Recent studies have highlighted the significant impact of neutrophils on tumor occurrence and progression, as well as their crucial role in the pancreatic cancer tumor microenvironment. Neutrophil infiltration plays a critical role in the progression and prognosis of pancreatic cancer. Neutrophils contribute to pancreatic cancer liver metastasis through various mechanisms, including angiogenesis, immune suppression, immune evasion, and epithelial-mesenchymal transition (EMT). Therefore, targeting neutrophils holds promise as an important therapeutic strategy for inhibiting pancreatic cancer liver metastasis. This article provides a summary of research findings on the involvement of neutrophils in pancreatic cancer liver metastasis and analyzes their potential as therapeutic targets. This research may provide new insights for the treatment of pancreatic cancer and improve the prognosis of patients with this disease.


Asunto(s)
Neoplasias Hepáticas , Neutrófilos , Neoplasias Pancreáticas , Microambiente Tumoral , Humanos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Neutrófilos/inmunología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/secundario , Animales , Microambiente Tumoral/inmunología , Transición Epitelial-Mesenquimal , Infiltración Neutrófila/efectos de los fármacos , Terapia Molecular Dirigida
5.
Biomed Pharmacother ; 178: 117269, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39137654

RESUMEN

Excessive neutrophil infiltration into the tumor microenvironment (TME) is an important factor that contributes to tumor overgrowth and limited immunotherapy efficacy. Neutrophils activate various receptors involved in tumor progression, while suppressing the infiltration and activity of cytotoxic T cells and creating optimal conditions for tumor growth. Therefore, the appropriate control of neutrophil infiltration is an effective strategy for tumor treatment. In the present study, 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) inhibited tumor overgrowth by suppressing excessive neutrophil infiltration, resulting in >74.97 % reduction in tumor size in a Lewis lung carcinoma (LLC-1) mouse model. All subjects in the positive control group died during the 90-day survival period, whereas only four subjects in the PLAG treatment group survived. PLAG had a significantly higher tumor growth inhibitory effect and survival rate than other neutrophil infiltration-targeting inhibitors (e.g., Navarixin, lymphocyte antigen 6 complex locus G6D antibody [aLy6G]). The ability of PLAG to regulate neutrophil infiltration and inhibit tumor growth depends on thioredoxin-interacting protein (TXNIP). In tumors lacking TXNIP expression, PLAG failed to control neutrophil infiltration and infiltration-related factor release, and the inhibitory effect of PLAG on tumor growth was reduced. PLAG-mediated inhibition of neutrophil infiltration enhances the efficacy of immune checkpoint inhibitors (ICIs), increasing the antitumor efficacy and survival rate by 30 %. In conclusion, PLAG could be a novel alternative to anti-tumor drugs that effectively targets excessive neutrophil infiltration into cancer tissues.


Asunto(s)
Carcinoma Pulmonar de Lewis , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Ratones Endogámicos C57BL , Infiltración Neutrófila , Microambiente Tumoral , Animales , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/inmunología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/inmunología , Ratones , Infiltración Neutrófila/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Microambiente Tumoral/efectos de los fármacos , Diglicéridos/farmacología , Línea Celular Tumoral , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Modelos Animales de Enfermedad , Masculino , Antineoplásicos/farmacología , Glicéridos
6.
Nat Commun ; 15(1): 7023, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174529

RESUMEN

Neutrophil infiltration and subsequent extracellular trap formation (NETosis) is a contributing factor in sterile inflammation. Furthermore, neutrophil extracellular traps (NETs) are prothrombotic, as they provide a scaffold for platelets and red blood cells to attach to. In circulation, neutrophils are constantly exposed to hemodynamic forces such as shear stress, which in turn regulates many of their biological functions such as crawling and NETosis. However, the mechanisms that mediate mechanotransduction in neutrophils are not fully understood. In this study, we demonstrate that shear stress induces NETosis, dependent on the shear stress level, and increases the sensitivity of neutrophils to NETosis-inducing agents such as adenosine triphosphate and lipopolysaccharides. Furthermore, shear stress increases intracellular calcium levels in neutrophils and this process is mediated by the mechanosensitive ion channel Piezo1. Activation of Piezo1 in response to shear stress mediates calpain activity and cytoskeleton remodeling, which consequently induces NETosis. Thus, activation of Piezo1 in response to shear stress leads to a stepwise sequence of cellular events that mediates NETosis and thereby places neutrophils at the centre of localized inflammation and prothrombotic effects.


Asunto(s)
Calcio , Trampas Extracelulares , Canales Iónicos , Mecanotransducción Celular , Neutrófilos , Estrés Mecánico , Neutrófilos/metabolismo , Canales Iónicos/metabolismo , Canales Iónicos/genética , Humanos , Trampas Extracelulares/metabolismo , Calcio/metabolismo , Adenosina Trifosfato/metabolismo , Calpaína/metabolismo , Lipopolisacáridos/farmacología , Citoesqueleto/metabolismo , Infiltración Neutrófila , Inflamación/metabolismo
7.
Nat Commun ; 15(1): 6718, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39112467

RESUMEN

Psoriasis is a multifactorial, chronic inflammatory skin disease with unresolved questions on its primary events. Iron overload has been described in the epidermis of psoriasis patients, but its relevance remains unknown. We found that the key iron regulatory hormone hepcidin was highly expressed in the epidermis of psoriasis patients, especially the pustular variants resistant to treatments. In a murine model of acute skin inflammation, keratinocyte-derived hepcidin was required for iron retention in keratinocytes, leading to hyperproliferation of the epidermal layer and neutrophil recruitment, two main features of psoriatic skin lesions. Keratinocytes overexpressing hepcidin were sufficient to elicit these psoriasiform features in a transgenic mouse model. Furthermore, transcriptome analysis of these keratinocytes revealed canonical pathways found in human psoriasis, pointing to a causal role for hepcidin in the pathogenesis of the disease. Altogether, our data suggest that hepcidin could be an actionable target for skin psoriasis treatment, in addition to current therapeutics, or targeted as maintenance therapy during remission to prevent recurrence.


Asunto(s)
Proliferación Celular , Hepcidinas , Hierro , Queratinocitos , Ratones Transgénicos , Infiltración Neutrófila , Psoriasis , Piel , Hepcidinas/metabolismo , Hepcidinas/genética , Psoriasis/metabolismo , Psoriasis/patología , Animales , Queratinocitos/metabolismo , Humanos , Hierro/metabolismo , Ratones , Piel/metabolismo , Piel/patología , Modelos Animales de Enfermedad , Masculino , Femenino , Epidermis/metabolismo , Epidermis/patología , Ratones Endogámicos C57BL , Inflamación/metabolismo , Inflamación/patología
8.
PLoS Biol ; 22(8): e3002779, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39159283

RESUMEN

Neutrophils are the first immune cells to reach inflamed sites and contribute to the pathogenesis of chronic inflammatory skin diseases. Yet, little is known about the pattern of neutrophil infiltration in inflamed skin in vivo and the mechanisms mediating their recruitment. Here, we provide insight into the dynamics of neutrophil infiltration in skin in response to acute or repeated inflammatory stress, highlighting a novel keratinocyte- and keratin 17 (K17)-dependent mechanism that regulates neutrophil recruitment to inflamed skin. We used the phorbol ester TPA and UVB, alone or in combination, to induce sterile inflammation in mouse skin. A single TPA treatment results in a neutrophil influx in the dermis that peaks at 12 h and resolves within 24 h. A subsequent TPA treatment or a UVB challenge, when applied 24 h but not 48 h later, accelerates, amplifies, and prolongs neutrophil infiltration. This transient amplification response (TAR) is mediated by local signals in inflamed skin, can be recapitulated in ex vivo culture, and involves the K17-dependent sustainment of protein kinase Cα (PKCα) activity and release of chemoattractants by stressed keratinocytes. K17 binds RACK1, a scaffold protein essential for PKCα activity. The N-terminal head domain of K17 is crucial for its association with RACK1 and regulation of PKCα activity. Analysis of RNAseq data reveals a signature consistent with TAR and PKCα activation in inflammatory skin diseases. These findings uncover a novel, keratin-dependent mechanism that amplifies neutrophil recruitment in skin under stress, with direct implications for inflammatory skin disorders.


Asunto(s)
Queratina-17 , Queratinocitos , Infiltración Neutrófila , Neutrófilos , Proteína Quinasa C-alfa , Piel , Animales , Humanos , Masculino , Ratones , Inflamación/metabolismo , Inflamación/patología , Queratina-17/metabolismo , Queratina-17/genética , Queratinocitos/metabolismo , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Proteína Quinasa C-alfa/metabolismo , Receptores de Cinasa C Activada/metabolismo , Receptores de Cinasa C Activada/genética , Piel/metabolismo , Piel/patología , Estrés Fisiológico , Acetato de Tetradecanoilforbol/farmacología , Rayos Ultravioleta/efectos adversos
9.
Immun Inflamm Dis ; 12(8): e1343, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39092750

RESUMEN

The involvement of neutrophils in the lungs during the recovery phase of coronavirus disease 2019 (COVID-19) is not well defined mainly due to the limited accessibility of lung tissues from COVID-19 survivors. The lack of an appropriate small animal model has affected the development of effective therapeutic strategies. We here developed a long COVID mouse model to study changes in neutrophil phenotype and association with lung injury. Our data shows persistent neutrophil recruitment and neutrophil extracellular trap formation in the lungs for up to 30 days post-infection which correlates with lung fibrosis and inflammation.


Asunto(s)
COVID-19 , Modelos Animales de Enfermedad , Trampas Extracelulares , Pulmón , Neutrófilos , SARS-CoV-2 , Animales , Trampas Extracelulares/inmunología , COVID-19/inmunología , COVID-19/complicaciones , Ratones , Neutrófilos/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Pulmón/patología , Pulmón/inmunología , Pulmón/virología , Lesión Pulmonar/inmunología , Lesión Pulmonar/virología , Lesión Pulmonar/patología , Lesión Pulmonar/etiología , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Infiltración Neutrófila/inmunología , Humanos , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/etiología
10.
Pharmacol Rep ; 76(5): 1067-1078, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39179890

RESUMEN

BACKGROUND: Recently, we demonstrated that nicorandil inhibits mechanical allodynia induced by paclitaxel. In the present study, we evaluated the effect induced by nicorandil in a model of neuropathic pain induced by chronic constriction injury (CCI) in mice. We also investigated putative mechanisms underlying such an effect. METHODS: CCI was induced by three ligatures of the left sciatic nerve. Mechanical allodynia was evaluated by measuring the paw withdrawal threshold with an electronic von Frey apparatus. Concentrations of cytokines and myeloperoxidase activity were determined in the paw tissue, sciatic nerve, and dorsal root ganglia (DRG). RESULTS: Oral administration of two doses of nicorandil (150 mg/kg po), but not equimolar doses of nicotinamide or nicotinic acid, attenuated mechanical allodynia induced by CCI. Nicorandil activity was reduced by previous administration of glibenclamide (40 mg/kg) or naltrexone (5 mg/kg or 10 mg/kg). Two doses of nicorandil (150 mg/kg, po) reduced tumor necrosis factor-α, interleukin-1ß and interleukin-6, but not CXCL-1, concentrations in the paw tissue of CCI mice. Two doses of nicorandil (150 mg/kg, po) reduced concentrations of all these mediators in the sciatic nerve and DRG. Two doses of nicorandil (150 mg/kg, po) also reduced the myeloperoxidase activity in the paw tissue, sciatic nerve, and DRG. CONCLUSIONS: Nicorandil exhibits antiallodynic activity in a model of neuropathic pain induced by CCI. Inhibition of cytokines production and reduction of neutrophils recruitment in paw tissue, sciatic nerve, and DRG as well as activation of ATP-dependent potassium channels and opioidergic pathways, underlie nicorandil antiallodynic activity.


Asunto(s)
Citocinas , Modelos Animales de Enfermedad , Ganglios Espinales , Hiperalgesia , Canales KATP , Neuralgia , Nicorandil , Nervio Ciático , Animales , Nicorandil/farmacología , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Ganglios Espinales/metabolismo , Ganglios Espinales/efectos de los fármacos , Ratones , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo , Masculino , Citocinas/metabolismo , Canales KATP/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Gliburida/farmacología , Naltrexona/farmacología , Naltrexona/análogos & derivados , Peroxidasa/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Analgésicos/farmacología
11.
Front Immunol ; 15: 1403789, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39156897

RESUMEN

Streptococcus suis causes diseases in pigs and has emerged as a zoonotic agent. When infected, the host develops an exacerbated inflammation that can lead to septic shock and meningitis. Although neutrophils greatly infiltrate the lesions, their dynamics during S. suis infection remain poorly described. Moreover, very few studies reported on the production and role of a key factor in the regulation of neutrophils: the colony-stimulating granulocyte factor (G-CSF). In this study, we characterized the G-CSF-neutrophil axis in the pathogenesis of S. suis induced disease. Using a mouse model of S. suis infection, we first evaluated the recruitment of neutrophils and their activation profile by flow cytometry. We found that infection provokes a massive neutrophil recruitment from the bone marrow to the blood and spleen. In both compartments, neutrophils displayed multiple activation markers. In parallel, we observed high systemic levels of G-CSF, with a peak of production coinciding with that of neutrophil recruitment. We then neutralized the effects of G-CSF and highlighted its role in the release of neutrophils from the bone marrow to the blood. However, it did not affect bacteremia nor the cytokine storm induced by S. suis. In conclusion, systemic G-CSF induces the release of neutrophils from the bone marrow to the blood, but its role in inflammation or bacterial clearance seems to be compensated by unknown factors. A better understanding of the role of neutrophils and inflammatory mediators could lead to better strategies for controlling the infection caused by S. suis.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos , Infiltración Neutrófila , Neutrófilos , Infecciones Estreptocócicas , Streptococcus suis , Streptococcus suis/inmunología , Animales , Factor Estimulante de Colonias de Granulocitos/metabolismo , Infecciones Estreptocócicas/inmunología , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Infiltración Neutrófila/inmunología , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL
12.
Hepatol Commun ; 8(8)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39101776

RESUMEN

BACKGROUND: Cholestatic liver diseases induce local and systemic hypercoagulation, with neutrophil extracellular traps (NETs) serving as major drivers. These NETs have been linked to decreased liver function in patients with obstructive jaundice. However, the impact of NETs on liver hypercoagulation in cholestatic liver disease remains unknown. METHODS: We utilized bile duct ligation to create experimental mice and analyzed NETs formation in the liver. Fibrin deposition, tissue factor expression, and inflammation in the liver were visualized through western blot and immunohistochemical techniques. LSECs were incubated with isolated NETs, and we detected endothelial procoagulant activity using coagulation protein production assays and measuring endothelial permeability. In both in vivo and in vitro settings, DNase I was applied to clarify the effect of NETs on intrahepatic hypercoagulability, hepatotoxicity, LSEC, and macrophage activation or injury. RESULTS: Bile duct ligation mice exhibited significantly increased levels of NETs in liver tissue, accompanied by neutrophil infiltration, tissue necrosis, fibrin deposition, and thrombophilia compared to sham mice. Notably, NETs resulted in phosphatidylserine and tissue factor exposure on LSEC, enhancing coagulation Factor Xa and thrombin production. The enhanced procoagulant activity could be reversed by degrading NETs with DNase I. Additionally, NETs-induced permeability changes in LSECs, characterized by increased VE-cadherin expression and F-actin retraction, which could be rescued by DNase I. Meanwhile, NET formation is associated with KC activation and the formation of inflammatory factors. CONCLUSIONS: NETs promote intrahepatic activation of coagulation and inflammation, leading to liver tissue injury. Strategies targeting NET formation may offer a potential therapeutic approach for treating cholestatic liver disease.


Asunto(s)
Trampas Extracelulares , Hígado , Trombosis , Trampas Extracelulares/metabolismo , Animales , Ratones , Hígado/patología , Hígado/metabolismo , Trombosis/etiología , Trombosis/patología , Colestasis/patología , Colestasis/complicaciones , Modelos Animales de Enfermedad , Masculino , Tromboplastina/metabolismo , Trombofilia/etiología , Trombofilia/sangre , Fibrina/metabolismo , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Humanos , Infiltración Neutrófila , Factor Xa/metabolismo , Trombina/metabolismo
13.
Cell Commun Signal ; 22(1): 386, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39090602

RESUMEN

BACKGROUND: T-LAK cell-oriented protein kinase (TOPK) strongly promotes the malignant proliferation of cancer cells and is recognized as a promising biomarker of tumor progression. Psoriasis is a common inflammatory skin disease featured by excessive proliferation of keratinocytes. Although we have previously reported that topically inhibiting TOPK suppressed psoriatic manifestations in psoriasis-like model mice, the exact role of TOPK in psoriatic inflammation and the underlying mechanism remains elusive. METHODS: GEO datasets were analyzed to investigate the association of TOPK with psoriasis. Skin immunohistochemical (IHC) staining was performed to clarify the major cells expressing TOPK. TOPK conditional knockout (cko) mice were used to investigate the role of TOPK-specific deletion in IMQ-induced psoriasis-like dermatitis in mice. Flow cytometry was used to analyze the alteration of psoriasis-related immune cells in the lesional skin. Next, the M5-induced psoriasis cell model was used to identify the potential mechanism by RNA-seq, RT-RCR, and western blotting. Finally, the neutrophil-neutralizing antibody was used to confirm the relationship between TOPK and neutrophils in psoriasis-like dermatitis in mice. RESULTS: We found that TOPK levels were strongly associated with the progression of psoriasis. TOPK was predominantly increased in the epidermal keratinocytes of psoriatic lesions, and conditional knockout of TOPK in keratinocytes suppressed neutrophils infiltration and attenuated psoriatic inflammation. Neutrophils deletion by neutralizing antibody greatly diminished the suppressive effect of TOPK cko in psoriasis-like dermatitis in mice. In addition, topical application of TOPK inhibitor OTS514 effectively attenuated already-established psoriasis-like dermatitis in mice. Mechanismly, RNA-seq revealed that TOPK regulated the expression of some genes in the IL-17 signaling pathway, such as neutrophils chemokines CXCL1, CXCL2, and CXCL8. TOPK modulated the expression of neutrophils chemokines via activating transcription factors STAT3 and NF-κB p65 in keratinocytes, thereby promoting neutrophils infiltration and psoriasis progression. CONCLUSIONS: This study identified a crucial role of TOPK in psoriasis by regulating neutrophils infiltration, providing new insights into the pathogenesis of psoriasis.


Asunto(s)
Queratinocitos , Quinasas de Proteína Quinasa Activadas por Mitógenos , Infiltración Neutrófila , Psoriasis , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Imiquimod , Queratinocitos/patología , Queratinocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Neutrófilos/patología , Psoriasis/patología , Psoriasis/genética , Transducción de Señal , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Regulación hacia Arriba , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo
14.
J Pathol ; 264(2): 174-185, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39056146

RESUMEN

The CCL2-CCR2 axis is involved in lupus nephritis, however the precise roles in the mechanisms by which different pathological lesions develop after glomerular immune complex deposition remain elusive. Previously, we demonstrated that genetic CCR2 inhibition induced a histological switch from glomerular endocapillary hypercellularity to wire-loop lesions in murine lupus nephritis. This study aimed to clarify the CCL2-CCR2 axis-mediated cellular mechanism in the formation of these different pathological lesions. We injected MRL/lpr mouse-derived monoclonal IgG3 antibody-producing hybridomas, 2B11.3 or B1, into wild-type (WT) mice to selectively induce glomerular endocapillary hypercellularity or wire-loop lesions. The expression of chemokine and chemokine receptors was analyzed using RT-quantitative PCR and/or immunofluorescence. We found 2B11.3 caused glomerular endocapillary hypercellularity in WT mice with glomerular infiltration of larger numbers of CCR2-expressing macrophages and neutrophils phagocyting immune complex, whereas B1 induced wire-loop lesions. In glomerular endocapillary hypercellularity, CCL2 was identified as the ligand involved in the CCR2-positive cell infiltration; it was expressed by glomerular endothelial cells and macrophages. Notably, 2B11.3-induced glomerular endocapillary hypercellularity converted to wire-loop lesions with reduced glomerular macrophage and neutrophil infiltration in CCL2-deficient (Ccl2-/-) mice similarly observed in Ccr2-/- mice. Moreover, this histological conversion was also observed when both glomerular macrophage and neutrophil infiltration were inhibited in anti-Ly6G antibody-treated Ccr5-/- mice but not when only glomerular macrophage infiltration was inhibited in Ccr5-/- mice or when only glomerular neutrophil infiltration was inhibited in anti-Ly6G antibody-treated WT mice. In contrast, B1 injection caused wire-loop lesions in Ccl2-/- and Ccr2-/- mice, as observed in WT mice. Moreover, 2B11.3 induced CCL2 from glomerular endothelial cells to a larger extent than B1 when injected into Ccr2-/- mice. In conclusion, the CCL2-CCR2 axis determines whether glomerular endocapillary hypercellularity or wire-loop lesions develop by regulating glomerular infiltration of phagocytic cells: macrophages and neutrophils. © 2024 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Quimiocina CCL2 , Glomérulos Renales , Nefritis Lúpica , Macrófagos , Receptores CCR2 , Animales , Nefritis Lúpica/patología , Nefritis Lúpica/inmunología , Nefritis Lúpica/metabolismo , Quimiocina CCL2/metabolismo , Receptores CCR2/metabolismo , Receptores CCR2/genética , Glomérulos Renales/patología , Glomérulos Renales/metabolismo , Glomérulos Renales/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Infiltración Neutrófila , Ratones Endogámicos MRL lpr , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , Femenino , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
15.
Stem Cell Res Ther ; 15(1): 225, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39075518

RESUMEN

BACKGROUND: This study explores the potential role of Thioredoxin-interacting protein (TXNIP) silencing in endothelial colony-forming cells (ECFCs) within the scope of age-related comorbidities and impaired vascular repair. We aim to elucidate the effects of TXNIP silencing on vasculogenic properties, paracrine secretion, and neutrophil recruitment under conditions of metabolic stress. METHODS: ECFCs, isolated from human blood cord, were transfected with TXNIP siRNA and exposed to a high glucose and ß-hydroxybutyrate (BHB) medium to simulate metabolic stress. We evaluated the effects of TXNIP silencing on ECFCs' functional and secretory responses under these conditions. Assessments included analyses of gene and protein expression profiles, vasculogenic properties, cytokine secretion and neutrophil recruitment both in vitro and in vivo. The in vivo effects were examined using a murine model of hindlimb ischemia to observe the physiological relevance of TXNIP modulation under metabolic disorders. RESULTS: TXNIP silencing did not mitigate the adverse effects on cell recruitment, vasculogenic properties, or senescence induced by metabolic stress in ECFCs. However, it significantly reduced IL-8 secretion and consequent neutrophil recruitment under these conditions. In a mouse model of hindlimb ischemia, endothelial deletion of TXNIP reduced MIP-2 secretion and prevented increased neutrophil recruitment induced by age-related comorbidities. CONCLUSIONS: Our findings suggest that targeting TXNIP in ECFCs may alleviate ischemic complications exacerbated by metabolic stress, offering potential clinical benefits for patients suffering from age-related comorbidities.


Asunto(s)
Proteínas Portadoras , Interleucina-8 , Infiltración Neutrófila , Estrés Fisiológico , Animales , Interleucina-8/metabolismo , Interleucina-8/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Humanos , Ratones , Infiltración Neutrófila/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/efectos de los fármacos , Isquemia/metabolismo , Isquemia/patología , ARN Interferente Pequeño/metabolismo , Tiorredoxinas/metabolismo , Tiorredoxinas/genética , Miembro Posterior/irrigación sanguínea , Ratones Endogámicos C57BL , Glucosa/metabolismo
16.
Front Immunol ; 15: 1411301, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39050842

RESUMEN

Ischemic heart disease (IHD) can trigger responses from the innate immune system, provoke aseptic inflammatory processes, and result in the recruitment and accumulation of neutrophils. Excessive recruitment of neutrophils is a potential driver of persistent cardiac inflammation. Once recruited, neutrophils are capable of secreting a plethora of inflammatory and chemotactic agents that intensify the inflammatory cascade. Additionally, neutrophils may obstruct microvasculature within the inflamed region, further augmenting myocardial injury in the context of IHD. Immune-related molecules mediate the recruitment process of neutrophils, such as immune receptors and ligands, immune active molecules, and immunocytes. Non-immune-related molecular pathways represented by pro-resolving lipid mediators are also involved in the regulation of NR. Finally, we discuss novel regulating strategies, including targeted intervention, agents, and phytochemical strategies. This review describes in as much detail as possible the upstream molecular mechanism and external intervention strategies for regulating NR, which represents a promising therapeutic avenue for IHD.


Asunto(s)
Isquemia Miocárdica , Infiltración Neutrófila , Neutrófilos , Humanos , Isquemia Miocárdica/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Infiltración Neutrófila/inmunología , Animales , Inflamación/inmunología , Inmunidad Innata , Mediadores de Inflamación/metabolismo
17.
Commun Biol ; 7(1): 887, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39033200

RESUMEN

Light serves as a crucial external zeitgeber for maintaining and restoring physiological homeostasis in most organisms. Disrupting of light rhythms often leads to abnormal immune function, characterized by excessive inflammatory responses. However, the underlying regulatory mechanisms behind this phenomenon remain unclear. To address this concern, we use in vivo imaging to establish inflammation models in zebrafish, allowing us to investigate the effects and underlying mechanisms of light disruption on neutrophil recruitment. Our findings reveal that under sustained light conditions (LL), neutrophil recruitment in response to caudal fin injury and otic vesicle inflammation is significantly increased. This is accompanied by elevated levels of histone (H3K18) lactylation and reactive oxygen species (ROS) content. Through ChIP-sequencing and ChIP‒qPCR analysis, we discover that H3K18 lactylation regulates the transcriptional activation of the duox gene, leading to ROS production. In turn, ROS further promote H3K18 lactylation, forming a positive feedback loop. This loop, driven by H3K18 lactylation-ROS, ultimately results in the over recruitment of neutrophils to inflammatory sites in LL conditions. Collectively, our study provides evidence of a mutual loop between histone lactylation and ROS, exacerbating neutrophil recruitment in light disorder conditions, emphasizing the significance of maintaining a proper light-dark cycle to optimize immune function.


Asunto(s)
Histonas , Luz , Infiltración Neutrófila , Especies Reactivas de Oxígeno , Pez Cebra , Animales , Pez Cebra/metabolismo , Histonas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Luz/efectos adversos , Neutrófilos/metabolismo , Neutrófilos/inmunología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Inflamación/metabolismo
18.
Circ Res ; 135(4): 503-517, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-38957990

RESUMEN

BACKGROUND: PANX1 (pannexin 1), a ubiquitously expressed ATP release membrane channel, has been shown to play a role in inflammation, blood pressure regulation, and myocardial infarction. However, the possible role of PANX1 in cardiomyocytes in the progression of heart failure has not yet been investigated. METHOD: We generated a novel mouse line with constitutive deletion of PANX1 in cardiomyocytes (Panx1MyHC6). RESULTS: PANX1 deletion in cardiomyocytes had no effect on unstressed heart function but increased the glycolytic metabolism and resulting glycolytic ATP production, with a concurrent decrease in oxidative phosphorylation, both in vivo and in vitro. In vitro, treatment of H9c2 (H9c2 rat myoblast cell line) cardiomyocytes with isoproterenol led to PANX1-dependent release of ATP and Yo-Pro-1 uptake, as assessed by pharmacological blockade with spironolactone and siRNA-mediated knockdown of PANX1. To investigate nonischemic heart failure and the preceding cardiac hypertrophy, we administered isoproterenol, and we demonstrated that Panx1MyHC6 mice were protected from systolic and diastolic left ventricle volume increases as a result of cardiomyocyte hypertrophy. Moreover, we found that Panx1MyHC6 mice showed decreased isoproterenol-induced recruitment of immune cells (CD45+), particularly neutrophils (CD11b+ [integrin subunit alpha M], Ly6g+ [lymphocyte antigen 6 family member G]), to the myocardium. CONCLUSIONS: Together, these data demonstrate that PANX1 deficiency in cardiomyocytes increases glycolytic metabolism and protects against cardiac hypertrophy in nonischemic heart failure at least in part by reducing immune cell recruitment. Our study implies PANX1 channel inhibition as a therapeutic approach to ameliorate cardiac dysfunction in patients with heart failure.


Asunto(s)
Conexinas , Glucólisis , Miocitos Cardíacos , Proteínas del Tejido Nervioso , Infiltración Neutrófila , Animales , Conexinas/genética , Conexinas/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Isoproterenol/farmacología , Cardiomegalia/metabolismo , Cardiomegalia/genética , Cardiomegalia/patología , Ratones Endogámicos C57BL , Línea Celular , Masculino , Adenosina Trifosfato/metabolismo , Ratones Noqueados , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología
19.
J Am Heart Assoc ; 13(15): e034492, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39028040

RESUMEN

BACKGROUND: Venous thromboembolism is associated with endothelial cell activation that contributes to the inflammation-dependent activation of the coagulation system. Cellular damage is associated with the release of different species of extracellular RNA (eRNA) involved in inflammation and coagulation. TLR3 (toll-like receptor 3), which recognizes (viral) single-stranded or double-stranded RNAs and self-RNA fragments, might be the receptor of these species of eRNA during venous thromboembolism. Here, we investigate how the TLR3/eRNA axis contributes to venous thromboembolism. METHODS AND RESULTS: Thrombus formation and size in wild-type and TLR3 deficient (-/-) mice were monitored by ultrasonography after venous thrombosis induction using the ferric chloride and stasis models. Mice were treated with RNase I, with polyinosinic-polycytidylic acid, a TLR3 agonist, or with RNA extracted from murine endothelial cells. Gene expression and signaling pathway activation were analyzed in HEK293T cells overexpressing TLR3 in response to eRNA or in human umbilical vein endothelial cells transfected with a small interference RNA against TLR3. Plasma clot formation on treated human umbilical vein endothelial cells was analyzed. Thrombosis exacerbated eRNA release in vivo and increased eRNA content within the thrombus. RNase I treatment reduced thrombus size compared with vehicle-treated mice (P<0.05). Polyinosinic-polycytidylic acid and eRNA treatments increased thrombus size in wild-type mice (P<0.01 and P<0.05), but not in TLR3-/- mice, by reinforcing neutrophil recruitment (P<0.05). Mechanistically, TLR3 activation in endothelial cells promotes CXCL5 (C-X-C motif chemokine 5) secretion (P<0.001) and NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation (P<0.05). Finally, eRNA triggered plasma clot formation in vitro (P<0.01). CONCLUSIONS: We show that eRNA and TLR3 activation enhance venous thromboembolism through neutrophil recruitment possibly through secretion of CXCL5, a potent neutrophil chemoattractant.


Asunto(s)
Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Receptor Toll-Like 3 , Trombosis de la Vena , Animales , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 3/genética , Trombosis de la Vena/metabolismo , Trombosis de la Vena/genética , Trombosis de la Vena/patología , Humanos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Transducción de Señal , Células HEK293 , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/patología , Neutrófilos/metabolismo , ARN/genética , Masculino , Ratones , Poli I-C/farmacología , Coagulación Sanguínea
20.
CNS Neurosci Ther ; 30(7): e14825, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38954749

RESUMEN

AIMS: Ischemic stroke remains a challenge in medical research because of the limited treatment options. Recombinant human tissue plasminogen activator (rtPA) is the primary treatment for recanalization. However, nearly 50% of the patients experience complications that result in ineffective reperfusion. The precise factors contributing to ineffective reperfusion remain unclear; however, recent studies have suggested that immune cells, notably neutrophils, may influence the outcome of rtPA thrombolysis via mechanisms such as the formation of neutrophil extracellular traps. This study aimed to explore the nonthrombolytic effects of rtPA on neutrophils and highlight their contribution to ineffective reperfusion. METHODS: We evaluated the effects of rtPA treatment on middle cerebral artery occlusion in rats. We also assessed neutrophil infiltration and activation after rtPA treatment in vitro and in vivo in a small cohort of patients with massive cerebral ischemia (MCI). RESULTS: rtPA increased neutrophil infiltration into the brain microvessels and worsened blood-brain barrier damage during ischemia. It also increased the neutrophil counts of the patients with MCI. CONCLUSION: Neutrophils play a crucial role in promoting ischemic injury and blood-brain barrier disruption, making them potential therapeutic targets.


Asunto(s)
Fibrinolíticos , Neutrófilos , Proteínas Recombinantes , Activador de Tejido Plasminógeno , Activador de Tejido Plasminógeno/farmacología , Activador de Tejido Plasminógeno/uso terapéutico , Animales , Humanos , Masculino , Neutrófilos/efectos de los fármacos , Ratas , Proteínas Recombinantes/farmacología , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Ratas Sprague-Dawley , Anciano , Barrera Hematoencefálica/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Femenino , Infiltración Neutrófila/efectos de los fármacos , Persona de Mediana Edad , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/inmunología , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA