Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.459
Filtrar
1.
Open Vet J ; 14(8): 1794-1800, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39308706

RESUMO

Background: Natural product active ingredients are currently being studied rigorously worldwide and offer a viable substitute for traditional immunotherapy for various medical disorders. Aim: The objective of the study was to investigate the immunostimulatory properties of fucoidan in albino Wistar rats. Methods: For the current study, forty rats were divided into five groups of rats that were used in good condition. In-vivo experiments of fucoidan were carried out in Wistar albino rats, such as the cyclophosphamide-caused myelosuppression, the delayed-type hypersensitivity (DTH) response, the phagocytic activity, the haemagglutinating antibody (HA) titer, and the neutrophil adhesion test. Results: The phagocytic index increased significantly in response to Fucoidan in a dose-dependent manner, as well as enhanced DTH reaction, and HA titer caused by sheep red blood cells sheep red blood cells. Additionally, fucoidan decreased myelosuppression in rats after cyclophosphamide treatment and enhanced neutrophil adhesion with nylon fiber. Conclusion: These findings imply that fucoidan has immunostimulant properties and could potentially utilised to treat immune-depression diseases.


Assuntos
Adjuvantes Imunológicos , Ciclofosfamida , Imunidade Celular , Imunidade Humoral , Polissacarídeos , Ratos Wistar , Animais , Polissacarídeos/farmacologia , Polissacarídeos/administração & dosagem , Ratos , Imunidade Humoral/efeitos dos fármacos , Ciclofosfamida/farmacologia , Imunidade Celular/efeitos dos fármacos , Adjuvantes Imunológicos/farmacologia , Hipersensibilidade Tardia/imunologia , Hipersensibilidade Tardia/tratamento farmacológico , Fagocitose/efeitos dos fármacos , Masculino , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia
2.
Biomed Pharmacother ; 179: 117360, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39232387

RESUMO

Macrophages undergo activation in response to multiple stimuli, including pathogens, growth factors and natural products. The inflammatory response and oxidative stress play critical roles in such macrophage activation. Some natural products reportedly promote immunoregulatory effects and the control of macrophage activation. Caryocar villosum (Cv), a native amazon plant, contains compounds that are an important source of molecules capable of macrophage activation. Herein, we demonstrate the immunomodulatory effects of oil obtained from Caryocar villosum (CvO) on macrophages. Macrophages were treated with varying concentrations of CvO, and resulting cellular morphological and functional changes were evaluated, including the production of nitric oxide (NO), reactive oxygen species (ROS), cytokines and phagocytic activity. Treatment of cells with 50 and 100 µg/mL CvO induced morphological and physiological alterations in the macrophages, such as increased cell surface and phagocytic activity. Additionally, treatment increased the productions of inflammatory cytokines (INF-γ, TNF-α, IL-6) and anti-inflammatory cytokines (IL-17 and IL-10) by macrophages, and significantly decreased ROS levels. In conclusion, these data suggest that, due to molecular diversity, CvO promoted an immunomodulatory effect on macrophages, mediated by an increased production of cytokines, and inhibition of ROS generation and phagocytic activity. Thus, CvO presents potential as a therapeutic agent for the treatment of inflammatory and non-inflammatory diseases.


Assuntos
Citocinas , Macrófagos , Óxido Nítrico , Fagocitose , Óleos de Plantas , Espécies Reativas de Oxigênio , Animais , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/imunologia , Citocinas/metabolismo , Fagocitose/efeitos dos fármacos , Óxido Nítrico/metabolismo , Óleos de Plantas/farmacologia , Células RAW 264.7 , Agentes de Imunomodulação/farmacologia , Agentes de Imunomodulação/isolamento & purificação , Fatores Imunológicos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Ericales/química
3.
Biomed Pharmacother ; 179: 117358, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39278188

RESUMO

Sodium houttuyfonate (SH), derived from the widely utilized natural herb Houttuynia cordata, exhibits an effective therapeutic effect on various diseases, including bacterial and fungal infections, especially the respiratory tract infection. Therefore, the anti-microbial mechanisms of SH may be different from the single-target action mechanism of conventional antibiotics, and further research is needed to clarify this. Firstly, we discovered that SH can effectively intervene in mouse lung infections by reducing bacterial load and acute inflammation response related to pneumonia caused by Pseudomonas aeruginosa. Interestingly, our results confirmed that SH has surface activity and can directly induce changes in the cell wall the shedding of surface lipopolysaccharide (LPS). Additionally, we found that SH-induced shedding of LPS can induce M1 polarization of macrophages in the early stage, leading to the production of corresponding polarization effector molecules. Subsequently, we discovered that SH-induced M1 polarization cells can effectively phagocytose and kill bacterial cells. The protein expression results indicated that SH can enhance the expression of M1 polarization pathway of TLR4/MyD88/NF-κB during the initial phase of macrophage and pathogen interaction. In summary, our results imply that SH could directly induce the shedding of P. aeruginosa LPS in a surfactant-like manner. Afterwards, the SH induced abscisic LPS can initiate the TLR4/MyD88/NF-κB immune pathway to trigger the M1 polarization of macrophages, which might intervene the P. aeruginosa-caused acute lung infection at early stage. Based on these findings, we attempted to coin the term "immune feedback eradication mechanism against pathogen of natural product" to describe this potent antimicrobial mechanism of SH.


Assuntos
Lipopolissacarídeos , Macrófagos , Pseudomonas aeruginosa , Sulfitos , Animais , Lipopolissacarídeos/farmacologia , Camundongos , Pseudomonas aeruginosa/efeitos dos fármacos , Sulfitos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Receptor 4 Toll-Like/metabolismo , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Alcanos/farmacologia , Células RAW 264.7 , Camundongos Endogâmicos C57BL , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/tratamento farmacológico , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Fagocitose/efeitos dos fármacos , Antibacterianos/farmacologia , Transdução de Sinais/efeitos dos fármacos
4.
World J Microbiol Biotechnol ; 40(10): 322, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39283509

RESUMO

Staphylococcus aureus can develop antibiotic resistance and evade immune responses, causing infections in different body sites. However, the metabolic changes underlying this process are poorly understood. A variant strain, C1V, was derived from the parental strain C1 by exposing it to increasing concentrations of vancomycin in vitro. C1V exhibited a vancomycin-intermediate phenotype and physiological changes compared to C1. It showed higher survival rates than C1 when phagocytosed by Raw264.7 cells. Metabolomics analysis identified significant metabolic differences pre- and post-induction (C1 + SC1 vs. C1V + SC1V: 201 metabolites) as well as pre- and post-phagocytosis (C1 vs. SC1: 50 metabolites; C1V vs. SC1V: 95 metabolites). The variant strain had distinct morphological characteristics, decreased adhesion ability, impaired virulence, and enhanced resistance to phagocytosis compared to the parental strain. Differential metabolites may contribute to S. aureus ' resistance to antibiotics and phagocytosis, offering insights into potential strategies for altering vancomycin nonsusceptibility and enhancing phagocyte killing by manipulating bacterial metabolism.


Assuntos
Antibacterianos , Metabolômica , Fagocitose , Staphylococcus aureus , Vancomicina , Vancomicina/farmacologia , Camundongos , Animais , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Fagocitose/efeitos dos fármacos , Células RAW 264.7 , Antibacterianos/farmacologia , Virulência , Infecções Estafilocócicas/microbiologia , Testes de Sensibilidade Microbiana , Resistência a Vancomicina/genética , Metaboloma/efeitos dos fármacos , Aderência Bacteriana/efeitos dos fármacos , Adaptação Fisiológica
5.
Allergol Immunopathol (Madr) ; 52(5): 73-79, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39278854

RESUMO

This study examines the therapeutic effects of Shengmai Powder (SMP) on both in vitro and in vivo models of chronic obstructive pulmonary disease (COPD) and the underlying mechanisms. Cigarette smoke and cigarette extracts were used to create in vitro and in vivo models of COPD. ELISA was used to measure the levels of pro-inflammatory factors (IL-6, TNF-α, and IL-1ß) in mouse lung tissue and alveolar macrophages. Flow cytometry assessed the phagocytic capacity of alveolar macrophage. Western blotting was used to analyze the expression of RhoA, PPARγ, IκBα, p-IκBα, P65, and p-P65 in alveolar. The results show that SMP reversed the increased levels of pro-inflammatory factors (IL-6, TNF-α, and IL-1ß) in mouse lung tissue and alveolar macrophages induced by cigarette smoke and cigarette extract. SMP also restored the decreased fluorescence intensity and RhoA levels in alveolar macrophages caused by cigarette extract. Additionally, SMP increased PPARγ expression and decreased IκBα and P65 phosphorylation in alveolar macrophages exposed to cigarette extract. Also, the effects of SMP were reversed by PPARγ inhibitors. The study concluded that SMP regulates alveolar macrophage phagocytic function through the PPAR-γ/NF-κB pathway, thereby improving the chronic inflammatory state of COPD.


Assuntos
Combinação de Medicamentos , Medicamentos de Ervas Chinesas , Macrófagos Alveolares , PPAR gama , Doença Pulmonar Obstrutiva Crônica , Animais , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/imunologia , PPAR gama/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Camundongos , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Modelos Animais de Doenças , Fagocitose/efeitos dos fármacos , Humanos , Masculino , Citocinas/metabolismo , Pós , Transdução de Sinais/efeitos dos fármacos , NF-kappa B/metabolismo
6.
Nat Commun ; 15(1): 8034, 2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39271657

RESUMO

Atherosclerosis is an inflammatory disorder responsible for cardiovascular disease. Reactivation of efferocytosis, the phagocytic removal of cells by macrophages, has emerged as a translational target for atherosclerosis. Systemic blockade of the key 'don't-eat-me' molecule, CD47, triggers the engulfment of apoptotic vascular tissue and potently reduces plaque burden. However, it also induces red blood cell clearance, leading to anemia. To overcome this, we previously developed a macrophage-specific nanotherapy loaded with a chemical inhibitor that promotes efferocytosis. Because it was found to be safe and effective in murine studies, we aimed to advance our nanoparticle into a porcine model of atherosclerosis. Here, we demonstrate that production can be scaled without impairing nanoparticle function. At an early stage of disease, we find our nanotherapy reduces apoptotic cell accumulation and inflammation in the atherosclerotic lesion. Notably, this therapy does not induce anemia, highlighting the translational potential of targeted macrophage checkpoint inhibitors.


Assuntos
Anemia , Aterosclerose , Antígeno CD47 , Modelos Animais de Doenças , Inflamação , Macrófagos , Nanopartículas , Fagocitose , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Nanopartículas/química , Antígeno CD47/metabolismo , Antígeno CD47/antagonistas & inibidores , Suínos , Inflamação/patologia , Fagocitose/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Humanos , Placa Aterosclerótica/patologia , Camundongos , Masculino
7.
J Alzheimers Dis ; 101(3): 787-796, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39240634

RESUMO

Background: Aggregated forms of the amyloid-ß (Aß) peptides which form protofibrils and fibrils in the brain are signatures of Alzheimer's disease (AD). Aggregates are also recognized by microglia, which in early phases may be protective and in later phases contribute to the pathology. We have identified several small molecules, decoys which interfere with Aß oligomerization and induce other aggregation trajectories leading to aggregated macrostructures which are non-toxic. Objective: This study investigates whether the small-molecule decoys affect microglial activation in terms of cytokine secretion and phagocytosis of Aß peptide. Methods: The effects of the decoys (NSC 69318, NSC 100873, NSC 16224) were analyzed in a model of human THP-1 monocytes differentiated to microglia-like cells. The cells were activated by Aß40 and Aß42 peptides, respectively, and after treatment with each decoy the secreted levels of pro-inflammatory cytokines and the Aß phagocytosis were analyzed. Results: NSC16224, which generates a double-stranded aggregate of thin protofibrils, was found to block Aß40- and Aß42-induced increase in microglial secretion of pro-inflammatory cytokines. NSC 69318, selective for neurotoxicity of Aß42, and NSC 100873 did not significantly reduce the microglial activation in terms of cytokine secretion. The uptake of Aß42 was not affected by anyone of the decoys. Conclusions: Our findings open the possibility that the molecular decoys of Aß aggregation may block microglial activation by Aß40 and Aß42 in addition to blocking neurotoxicity as shown previously.


Assuntos
Peptídeos beta-Amiloides , Microglia , Fragmentos de Peptídeos , Fagocitose , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Microglia/efeitos dos fármacos , Microglia/metabolismo , Humanos , Fagocitose/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Fragmentos de Peptídeos/farmacologia , Citocinas/metabolismo , Células THP-1
8.
Nat Commun ; 15(1): 8203, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39313508

RESUMO

Cytotherapy is a strategy to deliver modified cells to a diseased tissue, but targeting solid tumours remains challenging. Here we design macrophages, harbouring a surface glypican-3-targeting peptide and carrying a cargo to combat solid tumours. The anchored targeting peptide facilitates tumour cell recognition by the engineered macrophages, thus enhancing specific targeting and phagocytosis of tumour cells expressing glypican-3. These macrophages carry a cargo of the TLR7/TLR8 agonist R848 and INCB024360, a selective indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor, wrapped in C16-ceramide-fused outer membrane vesicles (OMV) of Escherichia coli origin (RILO). The OMVs facilitate internalization through caveolin-mediated endocytosis, and to maintain a suitable nanostructure, C16-ceramide induces membrane invagination and exosome generation, leading to the release of cargo-packed RILOs through exosomes. RILO-loaded macrophages exert therapeutic efficacy in mice bearing H22 hepatocellular carcinomas, which express high levels of glypican-3. Overall, we lay down the proof of principle for a cytotherapeutic strategy to target solid tumours and could complement conventional treatment.


Assuntos
Exossomos , Glipicanas , Macrófagos , Animais , Glipicanas/metabolismo , Exossomos/metabolismo , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Humanos , Linhagem Celular Tumoral , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/metabolismo , Modelos Animais de Doenças , Receptor 7 Toll-Like/metabolismo , Receptor 7 Toll-Like/agonistas , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Terapia Baseada em Transplante de Células e Tecidos/métodos , Sistemas de Liberação de Medicamentos , Fagocitose/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/metabolismo , Feminino
9.
ACS Nano ; 18(37): 25446-25464, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39240217

RESUMO

The alarming rise in global antimicrobial resistance underscores the urgent need for effective antibacterial drugs. Drawing inspiration from the bacterial-entrapment mechanism of human defensin 6, we have fabricated biomimetic peptide nanonets composed of multiple functional fragments for bacterial eradication. These biomimetic peptide nanonets are designed to address antimicrobial resistance challenges through a dual-approach strategy. First, the resulting nanofibrous networks trap bacteria and subsequently kill them by loosening the membrane structure, dissipating proton motive force, and causing multiple metabolic perturbations. Second, these trapped bacterial clusters reactivate macrophages to scavenge bacteria through enhanced chemotaxis and phagocytosis via the PI3K-AKT signaling pathway and ECM-receptor interaction. In vivo results have proven that treatment with biomimetic peptide nanonets can alleviate systemic bacterial infections without causing noticeable systemic toxicity. As anticipated, the proposed strategy can address stubborn infections by entrapping bacteria and awakening antibacterial immune responses. This approach might serve as a guide for the design of bioinspired materials for future clinical applications.


Assuntos
Antibacterianos , Materiais Biomiméticos , Macrófagos , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Macrófagos/metabolismo , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Antibacterianos/farmacologia , Antibacterianos/química , Humanos , Animais , Camundongos , Peptídeos/química , Peptídeos/farmacologia , Testes de Sensibilidade Microbiana , Staphylococcus aureus/efeitos dos fármacos , Células RAW 264.7 , Fagocitose/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos
10.
Immunohorizons ; 8(9): 652-667, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39259208

RESUMO

Bruton tyrosine kinase (BTK) is a kinase expressed by various immune cells and is often activated under proinflammatory states. Although the majority of BTK-related research has historically focused on B cells, understanding the role of BTK in non-B cell populations is critical given myeloid cells also express BTK at comparable levels. In this study, we investigated and compared how BTK inhibition in human and murine myeloid cells alters cell phenotype and function. All experiments were performed using two BTK inhibitors (evobrutinib and tolebrutinib) that are currently in late-stage clinical trials for the treatment of multiple sclerosis. Assays were performed to assess the impact of BTK inhibition on cytokine and microRNA expression, phagocytic capacity, and cellular metabolism. In all cells, both evobrutinib and tolebrutinib significantly decreased phosphorylated BTK and LPS-induced cytokine release. BTK inhibition also significantly decreased the oxygen consumption rate and extracellular acidification rate in myeloid cells, and significantly decreased phagocytosis in murine-derived cells, but not human macrophages. To further elucidate the mechanism, we also investigated the expression of microRNAs known to impact the function of myeloid cells. BTK inhibition resulted in an altered microRNA expression profile (i.e., decreased miR-155-5p and increased miR-223-3p), which is consistent with a decreased proinflammatory myeloid cell phenotype. In summary, these results provide further insights into the mechanism of action of BTK inhibitors in the context of immune-related diseases, while also highlighting important species-specific and cell-specific differences that should be considered when interpreting and comparing results between preclinical and human studies.


Assuntos
Tirosina Quinase da Agamaglobulinemia , Inflamação , MicroRNAs , Células Mieloides , Fagocitose , Tirosina Quinase da Agamaglobulinemia/metabolismo , Tirosina Quinase da Agamaglobulinemia/antagonistas & inibidores , Animais , Humanos , Camundongos , Fagocitose/efeitos dos fármacos , Células Mieloides/metabolismo , Células Mieloides/efeitos dos fármacos , Inflamação/metabolismo , Inflamação/tratamento farmacológico , MicroRNAs/metabolismo , MicroRNAs/genética , Inibidores de Proteínas Quinases/farmacologia , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Citocinas/metabolismo , Camundongos Endogâmicos C57BL
11.
Cell Death Dis ; 15(8): 625, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39223107

RESUMO

Parkinson's Disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the Substantia nigra pars compacta (SNpc). Apoptosis is thought to play a critical role in the progression of PD, and thus understanding the effects of antiapoptotic strategies is crucial for developing potential therapies. In this study, we developed a unique genetic model to selectively delete Casp3, the gene encoding the apoptotic protein caspase-3, in dopaminergic neurons (TH-C3KO) and investigated its effects in response to a subacute regime of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration, which is known to trigger apoptotic loss of SNpc dopaminergic neurons. We found that Casp3 deletion did not protect the dopaminergic system in the long term. Instead, we observed a switch in the cell death pathway from apoptosis in wild-type mice to necrosis in TH-C3KO mice. Notably, we did not find any evidence of necroptosis in our model or in in vitro experiments using primary dopaminergic cultures exposed to 1-methyl-4-phenylpyridinium in the presence of pan-caspase/caspase-8 inhibitors. Furthermore, we detected an exacerbated microglial response in the ventral mesencephalon of TH-C3KO mice in response to MPTP, which mimicked the microglia neurodegenerative phenotype (MGnD). Under these conditions, it was evident the presence of numerous microglial phagocytic cups wrapping around apparently viable dopaminergic cell bodies that were inherently associated with galectin-3 expression. We provide evidence that microglia exhibit phagocytic activity towards both dead and stressed viable dopaminergic neurons through a galectin-3-dependent mechanism. Overall, our findings suggest that inhibiting apoptosis is not a beneficial strategy for treating PD. Instead, targeting galectin-3 and modulating microglial response may be more promising approaches for slowing PD progression.


Assuntos
Apoptose , Caspase 3 , Neurônios Dopaminérgicos , Galectina 3 , Microglia , Necrose , Fagocitose , Animais , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Microglia/metabolismo , Microglia/patologia , Microglia/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Galectina 3/metabolismo , Galectina 3/genética , Caspase 3/metabolismo , Camundongos , Fagocitose/efeitos dos fármacos , Camundongos Knockout , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Camundongos Endogâmicos C57BL , Masculino
12.
J Agric Food Chem ; 72(37): 20432-20443, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39253862

RESUMO

For background, Hirsutella sinensis, the only anamorphic fungus considered an effective substitute for Cordyceps sinensis, possesses immunoregulatory properties. However, the specific mechanism underlying the immunoregulatory function of Hirsutella sinensis remains unclear. The purpose is to investigate the therapeutic effects of Hirsutella sinensis alcohol extract (HSAE) on immune dysregulation and elucidate the underlying mechanisms involved. For methods, we established inflammatory and immunosuppression models in vitro and in vivo to evaluate the bidirectional immunoregulatory function of HSAE via qRT-PCR and immunoblotting. We also studied its potential mechanism via RNA sequencing and transcriptional analysis. We further established M1 and M2 cell models to explore the effect of HSAE on M1/M2 polarization using qRT-PCR, immunoblotting, and flow cytometry. For results, our data demonstrated enhanced proliferation, phagocytosis, and antipathogenic activities of macrophages. Treatment with HSAE led to increases in the proportions of CD3+ and CD4+ immune cells in cyclophosphamide-induced immunosuppressed mice. Additionally, HSAE reduced the lipopolysaccharide (LPS)-induced expression of Il1b, Il6, Ifnb1, and Cxcl10 by inhibiting the activation of the NF-κB and MAPK pathways in vitro and improved mouse survival by reducing the proportion of M1/M2 macrophages in septic mice. Finally, we found that HSAE inhibited M1 polarization by decreasing the expression of iNOS and CD86 and promoted M2 polarization by increasing the expression of ARG1 and CD206. For conclusions, our study provides evidence that HSAE has the potential to enhance immune responses and suppress excessive inflammation. These effects were realized by modulating macrophage polarization, providing novel insights into the fundamental mechanism underlying the bidirectional immunomodulatory effect of HSAE.


Assuntos
Macrófagos , Animais , Camundongos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Humanos , Células RAW 264.7 , NF-kappa B/genética , NF-kappa B/imunologia , NF-kappa B/metabolismo , Camundongos Endogâmicos C57BL , Agentes de Imunomodulação/farmacologia , Agentes de Imunomodulação/química , Fagocitose/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Fatores Imunológicos/farmacologia , Fatores Imunológicos/química
13.
Int J Mol Sci ; 25(17)2024 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-39273195

RESUMO

Gwakhyangjeonggi-san (GJS) is a traditional herbal medicine used in East Asia for the treatment of symptoms involving lower intestinal abnormalities; however, the effects of GJS on innate immunity and its cellular mechanisms of action have not been elucidated. In this study, we assessed the immune-enhancing activity and underlying mechanisms of GJS using RAW 264.7 murine macrophages. The results showed that GJS treatment significantly increased the secretion of nitric oxide and cytokines and their mRNA expression in macrophage RAW 264.7 cells without causing cytotoxicity. GJS treatment also significantly increased the production of reactive oxygen species, as well as inducing phagocytic activity, adhesion function, and migration ability, all of which improved the immune response. In addition, GJS activated nuclear factor-κB by promoting the phosphorylation and degradation of inhibitor of nuclear factor-κB alpha. Furthermore, GJS markedly increased the phosphorylation of mitogen-activated protein kinase in RAW 264.7 cells. These findings indicate that GJS has potential value as a dietary supplement for strengthening immunity.


Assuntos
Macrófagos , NF-kappa B , Óxido Nítrico , Animais , Camundongos , Células RAW 264.7 , NF-kappa B/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/imunologia , Óxido Nítrico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Citocinas/metabolismo , Fagocitose/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Extratos Vegetais/farmacologia , Fosforilação/efeitos dos fármacos
14.
J Neuroinflammation ; 21(1): 218, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39227967

RESUMO

Alzheimer's disease (AD) is the leading form of dementia, characterized by the accumulation and aggregation of amyloid in brain. Transient receptor potential vanilloid 2 (TRPV2) is an ion channel involved in diverse physiopathological processes, including microglial phagocytosis. Previous studies suggested that cannabidiol (CBD), an activator of TRPV2, improves microglial amyloid-ß (Aß) phagocytosis by TRPV2 modulation. However, the molecular mechanism of TRPV2 in microglial Aß phagocytosis remains unknown. In this study, we aimed to investigate the involvement of TRPV2 channel in microglial Aß phagocytosis and the underlying mechanisms. Utilizing human datasets, mouse primary neuron and microglia cultures, and AD model mice, to evaluate TRPV2 expression and microglial Aß phagocytosis in both in vivo and in vitro. TRPV2 was expressed in cortex, hippocampus, and microglia.Cannabidiol (CBD) could activate and sensitize TRPV2 channel. Short-term CBD (1 week) injection intraperitoneally (i.p.) reduced the expression of neuroinflammation and microglial phagocytic receptors, but long-term CBD (3 week) administration (i.p.) induced neuroinflammation and suppressed the expression of microglial phagocytic receptors in APP/PS1 mice. Furthermore, the hyper-sensitivity of TRPV2 channel was mediated by tyrosine phosphorylation at the molecular sites Tyr(338), Tyr(466), and Tyr(520) by protein tyrosine kinase JAK1, and these sites mutation reduced the microglial Aß phagocytosis partially dependence on its localization. While TRPV2 was palmitoylated at Cys 277 site and blocking TRPV2 palmitoylation improved microglial Aß phagocytosis. Moreover, it was demonstrated that TRPV2 palmitoylation was dynamically regulated by ZDHHC21. Overall, our findings elucidated the intricate interplay between TRPV2 channel regulated by tyrosine phosphorylation/dephosphorylation and cysteine palmitoylation/depalmitoylation, which had divergent effects on microglial Aß phagocytosis. These findings provide valuable insights into the underlying mechanisms linking microglial phagocytosis and TRPV2 sensitivity, and offer potential therapeutic strategies for managing AD.


Assuntos
Peptídeos beta-Amiloides , Lipoilação , Camundongos Transgênicos , Microglia , Fagocitose , Canais de Cátion TRPV , Tirosina , Animais , Camundongos , Microglia/metabolismo , Microglia/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo , Peptídeos beta-Amiloides/metabolismo , Fagocitose/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Tirosina/metabolismo , Lipoilação/efeitos dos fármacos , Células Cultivadas , Doença de Alzheimer/metabolismo , Canabidiol/farmacologia , Camundongos Endogâmicos C57BL , Canais de Cálcio
15.
Colloids Surf B Biointerfaces ; 244: 114182, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39216441

RESUMO

Abnormal amyloid ß-protein (Aß42) fibrillation is a key event in Alzheimer's disease (AD), and photodynamic therapy (PDT) possesses great potential in modulating Aß42 self-assembly. However, the poor blood-brain barrier (BBB) penetration, low biocompatibility, and limited tissue penetration depth of existing photosensitizers limit the progress of photo-oxidation strategies. In this paper, novel indocyanine green-modified graphene quantum dot nano-assemblies (NBGQDs-ICGs) were synthesized based on a molecular assembly strategy of electrostatic interactions for PDT inhibition of Aß42 self-assembly process and decomposition of preformed fibrils under near-infrared light. Combining the small-size structure of graphene quantum dots and the near-infrared light-responsive properties of ICGs, the NBGQDs-ICGs could achieve BBB penetration under 808 nm irradiation. More importantly, the neuroprotective mechanism of NBGQDs-ICG was studied for the first time by AFM, which effectively weakened the adhesion of Aß42 aggregates to the cell surface by blocking the interaction between Aß42 and the cell membrane, and restored the mechanical stability and adhesion of the neuron membrane. Meanwhile, NBGQDs-ICG promoted phagocytosis of Aß42 by microglia. In addition, the good biocompatibility and stability ensured the biosafety of NBGQDs-ICG in future clinical applications. We anticipate that such multifunctional nanocomponents may provide promising avenues for the development of novel AD inhibitors.


Assuntos
Peptídeos beta-Amiloides , Barreira Hematoencefálica , Pontos Quânticos , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/química , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Pontos Quânticos/química , Humanos , Animais , Grafite/química , Grafite/farmacologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Tamanho da Partícula , Verde de Indocianina/química , Verde de Indocianina/farmacologia , Fagocitose/efeitos dos fármacos , Carbono/química , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Propriedades de Superfície
16.
Transl Psychiatry ; 14(1): 338, 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39179543

RESUMO

Microglia, traditionally regarded as innate immune cells in the brain, drive neuroinflammation and synaptic dysfunctions in the early phases of Alzheimer disease (AD), acting upstream to Aß accumulation. Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its levels are significantly increased in neurodegenerative diseases, possibly contributing to the chronic inflammatory microglial response. On the other hand, CSF-1R inhibitors confer neuroprotection in preclinical models of neurodegenerative diseases. Here, we determined the effects of the CSF-1R inhibitor PLX3397 on the Aß-mediated synaptic alterations in ex vivo hippocampal slices. Electrophysiological findings show that PLX3397 rescues LTP impairment and neurotransmission changes induced by Aß. In addition, using confocal imaging experiments, we demonstrate that PLX3397 stimulates a microglial transition toward a phagocytic phenotype, which in turn promotes the clearance of Aß from glutamatergic terminals. We believe that the selective pruning of Aß-loaded synaptic terminals might contribute to the restoration of LTP and excitatory transmission alterations observed upon acute PLX3397 treatment. This result is in accordance with the mechanism proposed for CSF1R inhibitors, that is to eliminate responsive microglia and replace it with newly generated, homeostatic microglia, capable of promoting brain repair. Overall, our findings identify a connection between the rapid microglia adjustments and the early synaptic alterations observed in AD, possibly highlighting a novel disease-modifying target.


Assuntos
Aminopiridinas , Peptídeos beta-Amiloides , Hipocampo , Potenciação de Longa Duração , Microglia , Animais , Microglia/efeitos dos fármacos , Microglia/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Peptídeos beta-Amiloides/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Aminopiridinas/farmacologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Pirróis/farmacologia , Camundongos , Fagocitose/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Ácido Glutâmico/metabolismo
17.
Inflamm Res ; 73(10): 1747-1763, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39127870

RESUMO

OBJECTIVE AND DESIGN: The aim of this study was to investigate the effects of ethanol exposure on epigenetic markers in bone marrow (BM) and their impact on inflammatory response during Aspergillus fumigatus infection. RESULTS: Chronic ethanol exposure decreased H3K27me3 enrichment in the Il6 promoter region while increased H3K4me3 enrichment in Tnf. Chimeric mice were generated by transplanting BM from mice exposed to ethanol or water. Infection of ethanol-chimeric mice culminated in higher clinical scores, although there was no effect on mortality. However, previous chronic exposure to ethanol affects persistently the inflammatory response in lung tissue, demonstrated by increased lung damage, neutrophil accumulation and IL-6, TNF and CXCL2 production in ethanol-chimeric mice, resulting in a decreased neutrophil infiltration into the alveolar space. Neutrophil killing and phagocytosis were also significantly lower. Moreover, BM derived macrophages (BMDM) from ethanol-chimeric mice stimulated with A. fumigatus conidia exhibited higher levels of TNF, CXCL2 and IL-6 release and a higher killing activity. The Il6 promoter of BMDM from ethanol-chimeric mice exhibited a reduction in H3K27me3 enrichment, a finding also observed in BM donors exposed to ethanol. CONCLUSIONS: These evidences demonstrate that prior chronic alcohol exposure of bone-marrow modify immune effector cells functions impairing the inflammatory response during A. fumigatus infection. These findings highlight the persistent impact of chronic ethanol exposure on infectious disease outcomes.


Assuntos
Aspergilose , Aspergillus fumigatus , Etanol , Histonas , Interleucina-6 , Macrófagos , Neutrófilos , Regiões Promotoras Genéticas , Animais , Interleucina-6/genética , Interleucina-6/metabolismo , Neutrófilos/imunologia , Neutrófilos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Histonas/metabolismo , Aspergilose/imunologia , Camundongos Endogâmicos C57BL , Masculino , Pulmão/imunologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Fagocitose/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
18.
Cell Rep ; 43(8): 114613, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39116206

RESUMO

Leptomeningeal metastases (LMs) remain a devastating complication of non-small cell lung cancer (NSCLC), particularly following osimertinib resistance. We conducted single-cell RNA sequencing on cerebrospinal fluid (CSF) from EGFR-mutant NSCLC with central nervous system metastases. We found that macrophages of LMs displayed functional and phenotypic heterogeneity and enhanced immunosuppressive properties. A population of lipid-associated macrophages, namely RNASE1_M, were linked to osimertinib resistance and LM development, which was regulated by Midkine (MDK) from malignant epithelial cells. MDK exhibited significant elevation in both CSF and plasma among patients with LMs, with higher MDK levels correlating to poorer outcomes in an independent cohort. Moreover, MDK could promote macrophage M2 polarization with lipid metabolism and phagocytic function. Furthermore, malignant epithelial cells in CSF, particularly after resistance to osimertinib, potentially achieved immune evasion through CD47-SIRPA interactions with RNASE1_M. In conclusion, we revealed a specific subtype of macrophages linked to osimertinib resistance and LM development, providing a potential target to overcome LMs.


Assuntos
Acrilamidas , Compostos de Anilina , Carcinoma Pulmonar de Células não Pequenas , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pulmonares , Macrófagos , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/tratamento farmacológico , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Acrilamidas/farmacologia , Acrilamidas/uso terapêutico , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Animais , Camundongos , Linhagem Celular Tumoral , Feminino , Carcinomatose Meníngea/tratamento farmacológico , Carcinomatose Meníngea/patologia , Carcinomatose Meníngea/secundário , Metabolismo dos Lipídeos/efeitos dos fármacos , Antígeno CD47/metabolismo , Antígeno CD47/genética , Masculino , Fagocitose/efeitos dos fármacos , Receptores ErbB/metabolismo , Receptores ErbB/genética , Indóis , Pirimidinas
19.
Int Immunopharmacol ; 140: 112834, 2024 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-39116495

RESUMO

BACKGROUND: Atherosclerotic (AS) plaques require a dense necrotic core and a robust fibrous cap to maintain stability. While previous studies have indicated that the traditional Chinese medicine Huang Lian Jie Du Decoction (HLJDD) possesses the capability to stabilize AS plaques, the underlying mechanisms remain obscure. This study aims to delve deeper into the potential mechanisms by which HLJDD improves AS through an integrated research strategy. METHODS: Leveraging an AS model in ApoE-/- mice exposed to a high-fat diet (HFD), we scrutinized the therapeutic effects of HLJDD using microscopic observations, oil red O staining, HE staining and Masson staining. Employing comprehensive techniques of network pharmacology, bioinformatics, and molecular docking, we elucidated the mechanism by which HLJDD stabilizes AS plaques. In vitro experiments, utilizing ox-LDL-induced macrophages and apoptotic vascular smooth muscle cells (VSMCs), assessed the impact of HLJDD on efferocytosis and the role of SLC2A1. RESULTS: In vivo experiments showcased the efficacy of HLJDD in reducing the quantity of aortic plaques, diminishing lipid deposition, and enhancing plaque stability in AS mice. Employing network pharmacology and machine learning, we pinpointed SLC2A1 as a crucial regulatory target. Molecular docking further validated the binding of HLJDD components with SLC2A1. The experiments demonstrated a dose-dependent upregulation in SLC2A1 expression by HLJDD, amplifying efferocytosis. Importantly, this effect was reversed by the SLC2A1 inhibitor STF-31, highlighting the pivotal role of SLC2A1 as a target. CONCLUSION: The HLJDD can modulate macrophage efferocytosis by enhancing the expression levels of SLC2A1, thereby improving the stability of atherosclerotic plaques.


Assuntos
Medicamentos de Ervas Chinesas , Transportador de Glucose Tipo 1 , Macrófagos , Placa Aterosclerótica , Animais , Placa Aterosclerótica/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Camundongos , Masculino , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 1/genética , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL , Fagocitose/efeitos dos fármacos , Humanos , Simulação de Acoplamento Molecular , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Modelos Animais de Doenças , Apoptose/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Lipoproteínas LDL/metabolismo , Células RAW 264.7 , Camundongos Knockout para ApoE , Eferocitose
20.
Mar Drugs ; 22(8)2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39195484

RESUMO

This study was conducted to evaluate the potential anti-inflammatory and immune-enhancement properties of lipids derived from Aptocyclus ventricosus eggs on RAW264.7 cells. Firstly, we determined the fatty acid compositions of A. ventricosus lipids by performing gas chromatography analysis. The results showed that A. ventricosus lipids contained saturated fatty acids (24.37%), monounsaturated fatty acids (20.90%), and polyunsaturated fatty acids (54.73%). They also contained notably high levels of DHA (25.91%) and EPA (22.05%) among the total fatty acids. Our results for the immune-associated biomarkers showed that A. ventricosus lipids had immune-enhancing effects on RAW264.7 cells. At the maximum dose of 300 µg/mL, A. ventricosus lipids generated NO (119.53%) and showed greater phagocytosis (63.69%) ability as compared with untreated cells. A. ventricosus lipids also upregulated the expression of iNOS, IL-1ß, IL-6, and TNF-α genes and effectively upregulated the phosphorylation of MAPK (JNK, p38, and ERK) and NF-κB p65, indicating that these lipids could activate the MAPK and NF-κB pathways to stimulate macrophages in the immune system. Besides their immune-enhancing abilities, A. ventricosus lipids significantly inhibited LPS-induced RAW264.7 inflammatory responses via the NF-κB and MAPK pathways. The results indicated that these lipids significantly reduced LPS-induced NO production, showing a decrease from 86.95% to 38.89%. Additionally, these lipids downregulated the expression of genes associated with the immune response and strongly suppressed the CD86 molecule on the cell surface, which reduced from 39.25% to 33.80%. Collectively, these findings imply that lipids extracted from A. ventricosus eggs might have biological immunoregulatory effects. Thus, they might be considered promising immunomodulatory drugs and functional foods.


Assuntos
NF-kappa B , Transdução de Sinais , Animais , Camundongos , Células RAW 264.7 , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Lipídeos , Macrófagos/efeitos dos fármacos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Anti-Inflamatórios/isolamento & purificação , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ovos , Fagocitose/efeitos dos fármacos , Ácidos Graxos/farmacologia , Óxido Nítrico/metabolismo , Citocinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA