Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.504
Filtrar
1.
FASEB J ; 38(9): e23638, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38713098

RESUMEN

Diabetic retinopathy (DR) is associated with ocular inflammation leading to retinal barrier breakdown, vascular leakage, macular edema, and vision loss. DR is not only a microvascular disease but also involves retinal neurodegeneration, demonstrating that pathological changes associated with neuroinflammation precede microvascular injury in early DR. Macrophage activation plays a central role in neuroinflammation. During DR, the inflammatory response depends on the polarization of retinal macrophages, triggering pro-inflammatory (M1) or anti-inflammatory (M2) activity. This study aimed to determine the role of macrophages in vascular leakage through the tight junction complexes of retinal pigment epithelium, which is the outer blood-retinal barrier (BRB). Furthermore, we aimed to assess whether interleukin-10 (IL-10), a representative M2-inducer, can decrease inflammatory macrophages and alleviate outer-BRB disruption. We found that modulation of macrophage polarization affects the structural and functional integrity of ARPE-19 cells in a co-culture system under high-glucose conditions. Furthermore, we demonstrated that intravitreal IL-10 injection induces an increase in the ratio of anti-inflammatory macrophages and effectively suppresses outer-BRB disruption and vascular leakage in a mouse model of early-stage streptozotocin-induced diabetes. Our results suggest that modulation of macrophage polarization by IL-10 administration during early-stage DR has a promising protective effect against outer-BRB disruption and vascular leakage. This finding provides valuable insights for early intervention in DR.


Asunto(s)
Barrera Hematorretinal , Diabetes Mellitus Experimental , Retinopatía Diabética , Interleucina-10 , Macrófagos , Ratones Endogámicos C57BL , Animales , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Barrera Hematorretinal/metabolismo , Barrera Hematorretinal/patología , Interleucina-10/metabolismo , Ratones , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/metabolismo , Masculino , Humanos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Estreptozocina , Activación de Macrófagos/efectos de los fármacos , Modelos Animales de Enfermedad , Polaridad Celular/efectos de los fármacos
2.
J Neuroinflammation ; 21(1): 115, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38698374

RESUMEN

BACKGROUND: Macrophages play a pivotal role in the regulation of Japanese encephalitis (JE), a severe neuroinflammation in the central nervous system (CNS) following infection with JE virus (JEV). Macrophages are known for their heterogeneity, polarizing into M1 or M2 phenotypes in the context of various immunopathological diseases. A comprehensive understanding of macrophage polarization and its relevance to JE progression holds significant promise for advancing JE control and therapeutic strategies. METHODS: To elucidate the role of NADPH oxidase-derived reactive oxygen species (ROS) in JE progression, we assessed viral load, M1 macrophage accumulation, and cytokine production in WT and NADPH oxidase 2 (NOX2)-deficient mice using murine JE model. Additionally, we employed bone marrow (BM) cell-derived macrophages to delineate ROS-mediated regulation of macrophage polarization by ROS following JEV infection. RESULTS: NOX2-deficient mice exhibited increased resistance to JE progression rather than heightened susceptibility, driven by the regulation of macrophage polarization. These mice displayed reduced viral loads in peripheral lymphoid tissues and the CNS, along with diminished infiltration of inflammatory cells into the CNS, thereby resulting in attenuated neuroinflammation. Additionally, NOX2-deficient mice exhibited enhanced JEV-specific Th1 CD4 + and CD8 + T cell responses and increased accumulation of M1 macrophages producing IL-12p40 and iNOS in peripheral lymphoid and inflamed extraneural tissues. Mechanistic investigations revealed that NOX2-deficient macrophages displayed a more pronounced differentiation into M1 phenotypes in response to JEV infection, thereby leading to the suppression of viral replication. Importantly, the administration of H2O2 generated by NOX2 was shown to inhibit M1 macrophage polarization. Finally, oral administration of the ROS scavenger, butylated hydroxyanisole (BHA), bolstered resistance to JE progression and reduced viral loads in both extraneural tissues and the CNS, along with facilitated accumulation of M1 macrophages. CONCLUSION: In light of our results, it is suggested that ROS generated by NOX2 play a role in undermining the control of JEV replication within peripheral extraneural tissues, primarily by suppressing M1 macrophage polarization. Subsequently, this leads to an augmentation in the viral load invading the CNS, thereby facilitating JE progression. Hence, our findings ultimately underscore the significance of ROS-mediated macrophage polarization in the context of JE progression initiated JEV infection.


Asunto(s)
Macrófagos , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 2 , Animales , Ratones , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/virología , NADPH Oxidasa 2/metabolismo , NADPH Oxidasa 2/genética , Encefalitis Japonesa/inmunología , Especies Reactivas de Oxígeno/metabolismo , Virus de la Encefalitis Japonesa (Especie) , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/virología , Polaridad Celular/efectos de los fármacos , Polaridad Celular/fisiología
3.
Biochem Biophys Res Commun ; 711: 149911, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38603832

RESUMEN

Macrophages play a crucial role in host response and wound healing, with M2 polarization contributing to the reduction of foreign-body reactions induced by the implantation of biomaterials and promoting tissue regeneration. Electrical stimulation (ES) and micropatterned substrates have a significant impact on the macrophage polarization. However, there is currently a lack of well-established cell culture platforms for studying the synergistic effects of these two factors. In this study, we prepared a graphene free-standing substrate with 20 µm microgrooves using capillary forces induced by water evaporation. Subsequently, we established an ES cell culture platform for macrophage cultivation by integrating a self-designed multi-well chamber cell culture device. We observed that graphene microgrooves, in combination with ES, significantly reduce cell spreading area and circularity. Results from immunofluorescence, ELISA, and flow cytometry demonstrate that the synergistic effect of graphene microgrooves and ES effectively promotes macrophage M2 phenotypic polarization. Finally, RNA sequencing results reveal that the synergistic effects of ES and graphene microgrooves inhibit the macrophage actin polymerization and the downstream PI3K signaling pathway, thereby influencing the phenotypic transition. Our results demonstrate the potential of graphene-based microgrooves and ES to synergistically modulate macrophage polarization, offering promising applications in regenerative medicine.


Asunto(s)
Estimulación Eléctrica , Grafito , Macrófagos , Grafito/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Animales , Ratones , Células RAW 264.7 , Polaridad Celular/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal
4.
Eur J Pharmacol ; 972: 176560, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38604543

RESUMEN

Obese asthma is a unique asthma phenotype that decreases sensitivity to inhaled corticosteroids, and currently lacks efficient therapeutic medication. Celastrol, a powerful bioactive substance obtained naturally from the roots of Tripterygium wilfordii, has been reported to possess the potential effect of weight loss in obese individuals. However, its role in the treatment of obese asthma is not fully elucidated. In the present study, diet-induced obesity (DIO) mice were used with or without ovalbumin (OVA) sensitization, the therapeutic effects of celastrol on airway hyperresponsiveness (AHR) and airway inflammation were examined. We found celastrol significantly decreased methacholine-induced AHR in obese asthma, as well as reducing the infiltration of inflammatory cells and goblet cell hyperplasia in the airways. This effect was likely due to the inhibition of M1-type alveolar macrophages (AMs) polarization and the promotion of M2-type macrophage polarization. In vitro, celastrol yielded equivalent outcomes in Lipopolysaccharide (LPS)-treated RAW264.7 macrophage cells, featuring a reduction in the expression of M1 macrophage makers (iNOS, IL-1ß, TNF-α) and heightened M2 macrophage makers (Arg-1, IL-10). Mechanistically, the PI3K/AKT signaling pathway has been implicated in these processes. In conclusion, we demonstrated that celastrol assisted in mitigating various parameters of obese asthma by regulating the balance of M1/M2 AMs polarization.


Asunto(s)
Asma , Macrófagos Alveolares , Obesidad , Triterpenos Pentacíclicos , Triterpenos , Animales , Asma/tratamiento farmacológico , Triterpenos Pentacíclicos/farmacología , Obesidad/tratamiento farmacológico , Obesidad/complicaciones , Ratones , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Triterpenos/farmacología , Triterpenos/uso terapéutico , Células RAW 264.7 , Inflamación/tratamiento farmacológico , Inflamación/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Hipersensibilidad Respiratoria/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Ratones Endogámicos C57BL , Ovalbúmina , Polaridad Celular/efectos de los fármacos
5.
Respir Res ; 25(1): 186, 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38678295

RESUMEN

BACKGROUND: Influenza A viruses (IAV) are extremely common respiratory viruses for the acute exacerbation of chronic obstructive pulmonary disease (AECOPD), in which IAV infection may further evoke abnormal macrophage polarization, amplify cytokine storms. Melatonin exerts potential effects of anti-inflammation and anti-IAV infection, while its effects on IAV infection-induced AECOPD are poorly understood. METHODS: COPD mice models were established through cigarette smoke exposure for consecutive 24 weeks, evaluated by the detection of lung function. AECOPD mice models were established through the intratracheal atomization of influenza A/H3N2 stocks in COPD mice, and were injected intraperitoneally with melatonin (Mel). Then, The polarization of alveolar macrophages (AMs) was assayed by flow cytometry of bronchoalveolar lavage (BAL) cells. In vitro, the effects of melatonin on macrophage polarization were analyzed in IAV-infected Cigarette smoking extract (CSE)-stimulated Raw264.7 macrophages. Moreover, the roles of the melatonin receptors (MTs) in regulating macrophage polarization and apoptosis were determined using MTs antagonist luzindole. RESULTS: The present results demonstrated that IAV/H3N2 infection deteriorated lung function (reduced FEV20,50/FVC), exacerbated lung damages in COPD mice with higher dual polarization of AMs. Melatonin therapy improved airflow limitation and lung damages of AECOPD mice by decreasing IAV nucleoprotein (IAV-NP) protein levels and the M1 polarization of pulmonary macrophages. Furthermore, in CSE-stimulated Raw264.7 cells, IAV infection further promoted the dual polarization of macrophages accompanied with decreased MT1 expression. Melatonin decreased STAT1 phosphorylation, the levels of M1 markers and IAV-NP via MTs reflected by the addition of luzindole. Recombinant IL-1ß attenuated the inhibitory effects of melatonin on IAV infection and STAT1-driven M1 polarization, while its converting enzyme inhibitor VX765 potentiated the inhibitory effects of melatonin on them. Moreover, melatonin inhibited IAV infection-induced apoptosis by suppressing IL-1ß/STAT1 signaling via MTs. CONCLUSIONS: These findings suggested that melatonin inhibited IAV infection, improved lung function and lung damages of AECOPD via suppressing IL-1ß/STAT1-driven macrophage M1 polarization and apoptosis in a MTs-dependent manner. Melatonin may be considered as a potential therapeutic agent for influenza virus infection-induced AECOPD.


Asunto(s)
Apoptosis , Subtipo H3N2 del Virus de la Influenza A , Melatonina , Enfermedad Pulmonar Obstructiva Crónica , Animales , Melatonina/farmacología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/virología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Ratones , Apoptosis/efectos de los fármacos , Células RAW 264.7 , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Ratones Endogámicos C57BL , Masculino , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Progresión de la Enfermedad , Polaridad Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología
6.
Brain Behav ; 14(4): e3487, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38648385

RESUMEN

INTRODUCTION: Demyelination is a key factor in axonal degeneration and neural loss, leading to disability in multiple sclerosis (MS) patients. Transforming growth factor beta activated kinase 1 (TAK1) is a critical molecule involved in immune and inflammatory signaling pathways. Knockout of microglia TAK1 can inhibit autoimmune inflammation of the brain and spinal cord and improve the outcome of MS. However, it is unclear whether inhibiting TAK1 can alleviate demyelination. METHODS: Eight-week-old male c57bl/6j mice were randomly divided into five groups: (a) the control group, (b) the group treated with cuprizone (CPZ) only, (c) the group treated with 5Z-7-Oxozaenol (OZ) only, and (d) the group treated with both cuprizone and 15 µg/30 µg OZ. Demyelination in the mice of this study was induced by administration of CPZ (ig) at a daily dose of 400 mg/kg for consecutive 5 weeks. OZ was intraperitoneally administered at mentioned doses twice a week, starting from week 3 after beginning cuprizone treatment. Histology, rotarod test, grasping test, pole test, Western blot, RT-PCR, and ELISA were used to evaluate corpus callosum demyelination, behavioral impairment, oligodendrocyte differentiation, TAK1 signaling pathway expression, microglia, and related cytokines. RESULTS: Our results demonstrated that OZ protected against myelin loss and behavior impairment caused by CPZ. Additionally, OZ rescued the loss of oligodendrocytes in CPZ-induced mice. OZ inhibited the activation of JNK, p65, and p38 pathways, transformed M1 polarized microglia into M2 phenotype, and increased brain-derived neurotrophic factor (BDNF) expression to attenuate demyelination in CPZ-treated mice. Furthermore, OZ reduced the expression of proinflammatory cytokines and increases anti-inflammatory cytokines in CPZ-treated mice. CONCLUSION: These findings suggest that inhibiting TAK1 may be an effective approach for treating demyelinating diseases.


Asunto(s)
Cuprizona , Enfermedades Desmielinizantes , Lactonas , Ratones Endogámicos C57BL , Microglía , Resorcinoles , Zearalenona/administración & dosificación , Animales , Cuprizona/farmacología , Microglía/efectos de los fármacos , Microglía/metabolismo , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/inducido químicamente , Ratones , Masculino , Quinasas Quinasa Quinasa PAM/metabolismo , Zearalenona/farmacología , Zearalenona/análogos & derivados , Polaridad Celular/efectos de los fármacos , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/patología , Cuerpo Calloso/metabolismo , Modelos Animales de Enfermedad
7.
Cell Mol Biol Lett ; 29(1): 62, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684954

RESUMEN

BACKGROUND: Enhancing angiogenesis may be an effective strategy to promote functional recovery after ischemic stroke. Inflammation regulates angiogenesis. Microglia are crucial cells that initiate inflammatory responses after various brain injuries. Long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) plays a role in regulating brain injury. This study aimed to explore the effects of NEAT1-regulated microglial polarization on the neovascularization capacity of cerebrovascular endothelial cells and the underlying molecular regulatory mechanisms. METHODS: Mouse cerebral arterial endothelial cells (mCAECs) were co-cultured with BV-2 cells in different groups using a Transwell system. NEAT1 expression levels were measured by fluorescence quantitative reverse transcription PCR. Levels of IL-1ß, IL-6, TNF-α, Arg-1, IL-4, and IL-10 were determined using ELISA. Expression levels of CD86 and CD163 were detected by immunofluorescence. The neovascularization capacity of mCAECs was assessed using CCK-8, Transwell, Transwell-matrigel, and tube formation assays. Label-free quantification proteomics was carried out to identify differentially expressed proteins. Protein levels were measured by Western blotting. RESULTS: NEAT1 overexpression induced M1 polarization in BV-2 cells, whereas NEAT1 knockdown blocked lipopolysaccharide-induced M1 polarization in microglia. NEAT1-overexpressing BV-2 cells suppressed the angiogenic ability of mCAECs, and NEAT1-knocking BV-2 cells promoted the angiogenic ability of mCAECs under lipopolysaccharide treatment. Label-free quantitative proteomic analysis identified 144 upregulated and 131 downregulated proteins that were induced by NEAT1 overexpression. The AMP-activated protein kinase (AMPK) signaling pathway was enriched in the Kyoto Encyclopedia of Genes and Genomes analysis of the differentially expressed proteins. Further verification showed that NEAT1 inactivated the AMPK signaling pathway. Moreover, the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide reversed the effect of NEAT1 on BV-2 polarization and the regulatory effect of NEAT1-overexpressing BV-2 cells on the angiogenic ability of mCAECs. CONCLUSIONS: NEAT1 inhibits the angiogenic activity of mCAECs by inducing M1 polarization of BV-2 cells through the AMPK signaling pathway. This study further clarified the impact and mechanism of NEAT1 on microglia and the angiogenic ability of cerebrovascular endothelial cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Células Endoteliales , Microglía , ARN Largo no Codificante , Transducción de Señal , Animales , Microglía/metabolismo , Microglía/efectos de los fármacos , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Arterias Cerebrales/metabolismo , Arterias Cerebrales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Línea Celular , Polaridad Celular/efectos de los fármacos
8.
Biomed Pharmacother ; 174: 116535, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38581923

RESUMEN

Studies have shown that Sacubitril/valsartan (Sac/Val) can reduce myocardial inflammation in myocarditis mice, in addition to its the recommended treatment of heart failure. However, the underlying mechanisms of Sac/Val in myocarditis remain unclear. C-type natriuretic peptide (CNP), one of the targeting natriuretic peptides of Sac/Val, was recently reported to exert cardio-protective and anti-inflammatory effects in cardiovascular systems. Here, we focused on circulating levels of CNP in patients with acute myocarditis (AMC) and whether Sac/Val modulates inflammation by targeting CNP in experimental autoimmune myocarditis (EAM) mice as well as LPS-induced RAW 264.7 cells and bone marrow derived macrophages (BMDMs) models. Circulating CNP levels were higher in AMC patients compared to healthy controls, and these levels positively correlated with the elevated inflammatory cytokines IL-6 and monocyte count. In EAM mice, Sac/Val alleviated myocardial inflammation while augmenting circulating CNP levels rather than BNP and ANP, accompanied by reduction in intracardial M1 macrophage infiltration and expression of inflammatory cytokines IL-1ß, TNF-α, and IL-6. Furthermore, Sac/Val inhibited CNP degradation and directly blunted M1 macrophage polarization in LPS-induced RAW 264.7 cells and BMDMs. Mechanistically, the effects might be mediated by the NPR-C/cAMP/JNK/c-Jun signaling pathway apart from NPR-B/cGMP/NF-κB pathway. In conclusion, Sac/Val exerts a protective effect in myocarditis by increasing CNP concentration and inhibiting M1 macrophages polarization.


Asunto(s)
Aminobutiratos , Compuestos de Bifenilo , Combinación de Medicamentos , Macrófagos , Miocarditis , Péptido Natriurético Tipo-C , Valsartán , Animales , Ratones , Miocarditis/tratamiento farmacológico , Miocarditis/metabolismo , Miocarditis/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Aminobutiratos/farmacología , Valsartán/farmacología , Células RAW 264.7 , Masculino , Humanos , Compuestos de Bifenilo/farmacología , Péptido Natriurético Tipo-C/farmacología , Tetrazoles/farmacología , Enfermedad Aguda , Modelos Animales de Enfermedad , Femenino , Citocinas/metabolismo , Citocinas/sangre , Ratones Endogámicos C57BL , Antiinflamatorios/farmacología , Polaridad Celular/efectos de los fármacos
9.
Eur J Pharmacol ; 972: 176523, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38552937

RESUMEN

The spinal cord microglia play a pivotal role in neuroinflammation and neuropathic pain (NP). Sodium tanshinone IIA sulfonate (STS), a derivative of tanshinone IIA, has anti-inflammatory and anti-hyperalgesic effects. However, its underlying mechanism in NP remains unclear. This study aimed to investigate the effect of STS and elucidate possible mechanisms in a rat model of spared nerve injury. In vivo experiments, STS and AG490 were administered intraperitoneally once daily for 14 consecutive days after surgery. The results showed that the expression of miR-125b-5p in the spinal dorsal horn was substantially reduced, whereas signal transducer and activator of transcription 3 (STAT3) signaling was increased. After treatment with STS, the mechanical thresholds, expression of miR-125b-5p, and microglial M2 marker such as Arg-1 in the spinal cord horn increased significantly, whereas multiple pro-inflammatory cytokines and apoptosis were significantly reduced. Moreover, STAT3 pathway-related proteins and expression of the microglial M1 marker, CD68, were appreciably inhibited. In vitro, lipopolysaccharide (LPS) was used to induce an inflammatory response in BV-2 microglial cells. STS pretreatment inhibited LPS-stimulated pro-inflammatory cytokine secretion, reduced STAT3 pathway related-proteins and apoptosis, increased miR-125b-5p and proopiomelanocortin expression, and enhanced microglia transformation from M1 to M2 phenotype in BV-2 cells. These effects were reversed after the inhibition of miR-125b-5p expression in BV-2 cells. A dual-luciferase reporter assay confirmed that STAT3 binds to miR-125b-5p. In summary, these results suggest that STS exerts anti-hyperalgesic and anti-neuroinflammatory effects in rats with NP possibly via the miR-125b-5p/STAT3 axis.


Asunto(s)
MicroARNs , Microglía , Neuralgia , Enfermedades Neuroinflamatorias , Fenantrenos , Ratas Sprague-Dawley , Factor de Transcripción STAT3 , Transducción de Señal , Animales , Factor de Transcripción STAT3/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Masculino , Fenantrenos/farmacología , Fenantrenos/uso terapéutico , Ratas , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Transducción de Señal/efectos de los fármacos , Ratones , Línea Celular , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Modelos Animales de Enfermedad , Polaridad Celular/efectos de los fármacos
10.
Apoptosis ; 29(5-6): 635-648, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38393643

RESUMEN

Patients with metastatic colorectal cancer often have poor outcomes, primarily due to hepatic metastasis. Colorectal cancer (CRC) cells have the ability to secrete cytokines and other molecules that can remodel the tumor microenvironment, facilitating the spread of cancer to the liver. Kupffer cells (KCs), which are macrophages in the liver, can be polarized to M2 type, thereby promoting the expression of adhesion molecules that aid in tumor metastasis. Our research has shown that huachanshu (with bufalin as the main active monomer) can effectively inhibit CRC metastasis. However, the underlying mechanism still needs to be thoroughly investigated. We have observed that highly metastatic CRC cells have a greater ability to induce M2-type polarization of Kupffer cells, leading to enhanced metastasis. Interestingly, we have found that inhibiting the expression of IL-6, which is highly expressed in the serum, can reverse this phenomenon. Notably, bufalin has been shown to attenuate the M2-type polarization of Kupffer cells induced by highly metastatic Colorectal cancer (mCRC) cells and down-regulate IL-6 expression, ultimately inhibiting tumor metastasis. In this project, our aim is to study how high mCRC cells induce M2-type polarization and how bufalin, via the SRC-3/IL-6 pathway, can inhibit CRC metastasis. This research will provide a theoretical foundation for understanding the anti-CRC effect of bufalin.


Asunto(s)
Bufanólidos , Neoplasias del Colon , Interleucina-6 , Macrófagos del Hígado , Neoplasias Hepáticas , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Bufanólidos/farmacología , Bufanólidos/uso terapéutico , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Humanos , Animales , Interleucina-6/metabolismo , Interleucina-6/genética , Neoplasias del Colon/patología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/genética , Línea Celular Tumoral , Ratones , Antineoplásicos/farmacología , Microambiente Tumoral/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Metástasis de la Neoplasia
11.
Adv Healthc Mater ; 13(12): e2304114, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38295299

RESUMEN

The skin serves as the body's outermost barrier and is the largest organ, providing protection not only to the body but also to various internal organs. Owing to continuous exposure to various external factors, it is susceptible to damage that can range from simple to severe, including serious types of wounds such as burns or chronic wounds. Macrophages play a crucial role in the entire wound-healing process and contribute significantly to skin regeneration. Initially, M1 macrophages infiltrate to phagocytose bacteria, debris, and dead cells in fresh wounds. As tissue repair is activated, M2 macrophages are promoted, reducing inflammation and facilitating restoration of the dermis and epidermis to regenerate the tissue. This suggests that extracellular matrix (ECM) promotes cell adhesion, proliferation, migrationand macrophage polarization. Among the numerous strategies, electrospinning is a versatile technique for obtaining ECM-mimicking structures with anisotropic and isotropic topologies of micro/nanofibers. Various electrospun biomaterials influence macrophage polarization based on their isotropic or anisotropic topologies. Moreover, these fibers possess a high surface-area-to-volume ratio, promoting the effective exchange of vital nutrients and oxygen, which are crucial for cell viability and tissue regeneration. Micro/nanofibers with diverse physical and chemical properties can be tailored to polarize macrophages toward skin regeneration and wound healing, depending on specific requirements. This review describes the significance of micro/nanostructures for activating macrophages and promoting wound healing.


Asunto(s)
Matriz Extracelular , Macrófagos , Nanofibras , Cicatrización de Heridas , Nanofibras/química , Cicatrización de Heridas/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Humanos , Animales , Anisotropía , Polaridad Celular/efectos de los fármacos , Piel/lesiones , Piel/metabolismo
12.
Cell Mol Immunol ; 21(1): 60-79, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38062129

RESUMEN

The main challenges in the use of immune checkpoint inhibitors (ICIs) are ascribed to the immunosuppressive tumor microenvironment and the lack of sufficient infiltration of activated CD8+ T cells. Transforming the tumor microenvironment (TME) from "cold" to "hot" and thus more likely to potentiate the effects of ICIs is a promising strategy for cancer treatment. We found that the selective BCL-2 inhibitor APG-2575 can enhance the antitumor efficacy of anti-PD-1 therapy in syngeneic and humanized CD34+ mouse models. Using single-cell RNA sequencing, we found that APG-2575 polarized M2-like immunosuppressive macrophages toward the M1-like immunostimulatory phenotype with increased CCL5 and CXCL10 secretion, restoring T-cell function and promoting a favorable immunotherapy response. Mechanistically, we demonstrated that APG-2575 directly binds to NF-κB p65 to activate NLRP3 signaling, thereby mediating macrophage repolarization and the activation of proinflammatory caspases and subsequently increasing CCL5 and CXCL10 chemokine production. As a result, APG-2575-induced macrophage repolarization could remodel the tumor immune microenvironment, thus improving tumor immunosuppression and further enhancing antitumor T-cell immunity. Multiplex immunohistochemistry confirmed that patients with better immunotherapeutic efficacy had higher CD86, p-NF-κB p65 and NLRP3 levels, accompanied by lower CD206 expression on macrophages. Collectively, these data provide evidence that further study on APG-2575 in combination with immunotherapy for tumor treatment is required.


Asunto(s)
Dioxanos , Inhibidores de Puntos de Control Inmunológico , Terapia de Inmunosupresión , Neoplasias Pulmonares , Proteína con Dominio Pirina 3 de la Familia NLR , Nitrobencenos , Proteínas Proto-Oncogénicas c-bcl-2 , Pirroles , Macrófagos Asociados a Tumores , Animales , Ratones , Dioxanos/farmacología , Dioxanos/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Nitrobencenos/farmacología , Nitrobencenos/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR/agonistas , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Pirroles/farmacología , Pirroles/uso terapéutico , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/metabolismo , Factor de Transcripción ReIA/metabolismo , Microambiente Tumoral/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Humanos , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos C57BL , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/metabolismo , Terapia de Inmunosupresión/métodos
13.
J Cancer Res Ther ; 19(6): 1560-1567, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38156922

RESUMEN

OBJECTIVE: This study aimed to evaluate the impact of an adenosine monophosphate-activated protein kinase (AMPK) agonist, metformin (MET), on the antitumor effects of macrophages and to determine the underlying mechanism involved in the process. MATERIALS AND METHODS: M0 macrophages were derived from phorbol-12-myristate-13-acetate-stimulated THP-1 cells. RESULTS: The levels of tumor necrosis factor-alpha (TNF-α) and human leukocyte antigen-DR (HLA-DR) were decreased in macrophages incubated with HCT116 cells, whereas those of arginase-1 (Arg-1), CD163, and CD206 were elevated; these effects were reversed by MET. The transfection of small interfering (si) RNA abrogated the influence of MET on the expression of the M1/M2 macrophage biomarkers. MET significantly suppressed the proliferation and migration abilities of HCT116 cells incubated with M0 macrophages; these actions were reversed by siRNA transfection against AMPK. The hypoxia-inducible factor 1-alpha (HIF-1α), phosphorylated protein kinase B (p-AKT), and phosphorylated mammalian target of rapamycin (p-mTOR) levels were reduced by the introduction of MET and promoted by siRNA transfection against AMPK. In addition, the levels of HIF-1α, p-AKT, and p-mTOR suppressed by MET were markedly increased following the transfection of siRNA against AMPK. CONCLUSION: These findings indicate that MET can repress the progression of colorectal cancer by transforming tumor-associated macrophages to the M1phenotype via inhibition of the HIF-1α and mTOR signaling pathways.


Asunto(s)
Neoplasias Colorrectales , Metformina , Transducción de Señal , Serina-Treonina Quinasas TOR , Macrófagos Asociados a Tumores , Metformina/farmacología , Metformina/uso terapéutico , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Macrófagos Asociados a Tumores/efectos de los fármacos , Células HCT116 , Polaridad Celular/efectos de los fármacos , Células THP-1 , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Técnicas de Silenciamiento del Gen
14.
Int J Mol Sci ; 24(13)2023 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-37446007

RESUMEN

Some chemoattractants and leukocytes such as M1 and M2 macrophages are known to be involved in the development of glomerulosclerosis during diabetic nephropathy (DN). In the course of diabetes, an altered and defective cellular metabolism leads to the increase in adenosine levels, and thus to changes in the polarity (M1/M2) of macrophages. MRS1754, a selective antagonist of the A2B adenosine receptor (A2BAR), attenuated glomerulosclerosis and decreased macrophage-myofibroblast transition in DN rats. Therefore, we aimed to investigate the effect of MRS1754 on the glomerular expression/secretion of chemoattractants, the intraglomerular infiltration of leukocytes, and macrophage polarity in DN rats. Kidneys/glomeruli of non-diabetic, DN, and MRS1754-treated DN rats were processed for transcriptomic analysis, immunohistopathology, ELISA, and in vitro macrophage migration assays. The transcriptomic analysis identified an upregulation of transcripts and pathways related to the immune system in the glomeruli of DN rats, which was attenuated using MRS1754. The antagonism of the A2BAR decreased glomerular expression/secretion of chemoattractants (CCL2, CCL3, CCL6, and CCL21), the infiltration of macrophages, and their polarization to M2 in DN rats. The in vitro macrophages migration induced by conditioned-medium of DN glomeruli was significantly decreased using neutralizing antibodies against CCL2, CCL3, and CCL21. We concluded that the pharmacological blockade of the A2BAR decreases the transcriptional expression of genes/pathways related to the immune response, protein expression/secretion of chemoattractants, as well as the infiltration of macrophages and their polarization toward the M2 phenotype in the glomeruli of DN rats, suggesting a new mechanism implicated in the antifibrotic effect of MRS1754.


Asunto(s)
Acetamidas , Antagonistas del Receptor de Adenosina A2 , Polaridad Celular , Factores Quimiotácticos , Nefropatías Diabéticas , Glomérulos Renales , Macrófagos , Purinas , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/inmunología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/genética , Factores Quimiotácticos/metabolismo , Polaridad Celular/efectos de los fármacos , Polaridad Celular/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Antagonistas del Receptor de Adenosina A2/farmacología , Receptor de Adenosina A2B , Acetamidas/farmacología , Purinas/farmacología , Animales , Ratas , Movimiento Celular/efectos de los fármacos , Masculino , Ratas Sprague-Dawley , Transcripción Genética/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Inmunidad/efectos de los fármacos , Inmunidad/genética
15.
J Biol Chem ; 299(6): 104779, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37142224

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is associated with an increased ratio of classically activated M1 macrophages/Kupffer cells to alternatively activated M2 macrophages, which plays an imperative role in the development and progression of NAFLD. However, little is known about the precise mechanism behind macrophage polarization shift. Here, we provide evidence regarding the relationship between the polarization shift in Kupffer cells and autophagy resulting from lipid exposure. High-fat and high-fructose diet supplementation for 10 weeks significantly increased the abundance of Kupffer cells with an M1-predominant phenotype in mice. Interestingly, at the molecular level, we also observed a concomitant increase in expression of DNA methyltransferases DNMT1 and reduced autophagy in the NAFLD mice. We also observed hypermethylation at the promotor regions of autophagy genes (LC3B, ATG-5, and ATG-7). Furthermore, the pharmacological inhibition of DNMT1 by using DNA hypomethylating agents (azacitidine and zebularine) restored Kupffer cell autophagy, M1/M2 polarization, and therefore prevented the progression of NAFLD. We report the presence of a link between epigenetic regulation of autophagy gene and macrophage polarization switch. We provide the evidence that epigenetic modulators restore the lipid-induced imbalance in macrophage polarization, therefore preventing the development and progression of NAFLD.


Asunto(s)
Autofagia , Polaridad Celular , Macrófagos , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Autofagia/efectos de los fármacos , Autofagia/genética , Dieta Alta en Grasa/efectos adversos , Dieta Occidental/efectos adversos , Epigénesis Genética/efectos de los fármacos , Hígado/citología , Hígado/fisiopatología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Azacitidina/farmacología , Azacitidina/uso terapéutico , Inhibidores Enzimáticos/farmacología , Metilación de ADN/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Células RAW 264.7 , Técnicas de Silenciamiento del Gen
16.
J Cell Physiol ; 238(3): 631-646, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36727620

RESUMEN

A common adverse response to the clinical use of glucocorticoids (GCs) is elevated intraocular pressure (IOP) which is a major risk factor for glaucoma. Elevated IOP arises due to impaired outflow of aqueous humor (AH) through the trabecular meshwork (TM). Although GC-induced changes in actin cytoskeletal dynamics, contractile characteristics, and cell adhesive interactions of TM cells are believed to influence AH outflow and IOP, the molecular mechanisms mediating changes in these cellular characteristics are poorly understood. Our studies focused on evaluating changes in the cytoskeletal and cytoskeletal-associated protein (cytoskeletome) profile of human TM cells treated with dexamethasone (Dex) using label-free mass spectrometric quantification, identified elevated levels of specific proteins known to regulate actin stress fiber formation, contraction, actin networks crosslinking, cell adhesion, and Wnt signaling, including LIMCH1, ArgBP2, CNN3, ITGBL1, CTGF, palladin, FAT1, DIAPH2, EPHA4, SIPA1L1, and GPC4. Several of these proteins colocalized with the actin cytoskeleton and underwent alterations in distribution profile in TM cells treated with Dex, and an inhibitor of Abl/Src kinases. Wnt/Planar Cell Polarity (PCP) signaling agonists-Wnt5a and 5b were detected prominently in the cytoskeletome fraction of TM cells, and studies using siRNA to suppress expression of glypican-4 (GPC4), a known modulator of the Wnt/PCP pathway revealed that GPC4 deficiency impairs Dex induced actin stress fiber formation, and activation of c-Jun N-terminal Kinase (JNK) and Rho kinase. Additionally, while Dex augmented, GPC4 deficiency suppressed the formation of actin stress fibers in TM cells in the presence of Dex and Wnt5a. Taken together, these results identify the GPC4-dependent Wnt/PCP signaling pathway as one of the crucial upstream regulators of Dex induced actin cytoskeletal reorganization and cell adhesion in TM cells, opening an opportunity to target the GPC4/Wnt/PCP pathway for treatment of ocular hypertension in glaucoma.


Asunto(s)
Actinas , Proteínas del Citoesqueleto , Citoesqueleto , Dexametasona , Glucocorticoides , Glipicanos , Malla Trabecular , Humanos , Actinas/metabolismo , Células Cultivadas , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Dexametasona/farmacología , Glaucoma/metabolismo , Glaucoma/patología , Glucocorticoides/farmacología , Glipicanos/deficiencia , Glipicanos/metabolismo , Presión Intraocular , Malla Trabecular/citología , Malla Trabecular/efectos de los fármacos , Malla Trabecular/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Citoesqueleto/metabolismo , Polaridad Celular/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo , Fibras de Estrés/efectos de los fármacos , Adhesión Celular/efectos de los fármacos
17.
Neuropharmacology ; 212: 109064, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35452626

RESUMEN

Microglia are immune cells in the central nervous system (CNS) that participate in response to pathological process after ischemic injury. Non-mitogenic fibroblast growth factor 1 (nmFGF1) is an effective neuroprotective factor that is also known as a metabolic regulator. The present study aimed to investigate the effects and mechanism of the neuroprotective ability of nmFGF1 on microglia in mice after photothrombosis (PT) stroke model, to determine whether it could ameliorate ischemic injury in stroke experiment. We discovered that the intranasal administration of nmFGF1 reduced infarct size and ameliorated neurological deficits in behavioral assessment by regulating the secretion of proinflammatory and anti-inflammatory cytokines. Furthermore, in the in vitro experiments, we found that nmFGF1 regulated the expression levels of proinflammatory and anti-inflammatory cytokines in oxygen-glucose deprivation (OGD) and lipopolysaccharide (LPS) stimulation. Evidence have shown that when nuclear factor erythroid 2-related factor 2 (Nfr2) is activated, it inhibits nuclear factor-kappa B (NF-κB) activation to alleviate inflammation. Interestingly, nmFGF1 treatment in vivo remarkably inhibited NF-κB pathway activation and activated Nrf2 pathway. In addition, nmFGF1 and NF-κB inhibitor (BAY11-7082) inhibited NF-κB pathway in LPS-stimulated BV2 microglia. Moreover, in LPS-stimulated BV2 microglia, the anti-inflammatory effect produced by nmFGF1 was knocked down by Nrf2 siRNA. These results indicate that nmFGF1 promoted functional recovery in experimental stroke by modulating microglia/macrophage-mediated neuroinflammation via Nrf2 and NF-κB signaling pathways, making nmFGF1 a potential agent against ischemic stroke.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos , Accidente Cerebrovascular Isquémico , Macrófagos , Microglía , Factor 2 Relacionado con NF-E2 , FN-kappa B , Accidente Cerebrovascular , Animales , Antiinflamatorios/farmacología , Polaridad Celular/efectos de los fármacos , Citocinas/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Microglía/metabolismo , Microglía/patología , Mitógenos/metabolismo , Mitógenos/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
18.
Mol Biol Rep ; 49(5): 3975-3986, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35166983

RESUMEN

BACKGROUND: The inflammatory response caused by microglia in the central nervous system plays an important role in Alzheimer's disease. Neuregulin-1 (NRG1) is a member of the neuregulin family and has been demonstrated to have anti-inflammatory properties. The relationship between NRG1, microglia phenotype and neuroinflammation remains unclear. MATERIALS AND METHODS: BV2 cells were used to examine the mechanism of NRG1 in regulating microglia polarization. Neuronal apoptosis, inflammatory factors TNF-α and iNOS, microglia polarization, ErbB4 and NF-κB p65 expression were assessed. RESULTS: We found that exogenous NRG1 treatment or overexpression improved microglial activity and reduced the secretion of the inflammatory factors TNF-α and iNOS in vitro. The expression of Bax in SH-SY5Y neuron cells incubated with medium collected from the NRG1 treatment group decreased. Additionally, our study showed that NRG1 treatment reduced the levels of the M1 microglia markers CD120 and iNOS and increased the levels of the M2 microglia markers CD206 and Arg-1. Furthermore, we observed that NRG1 treatment attenuated Aß-induced NF-κB activation and promoted the expression of p-ErbB4 and that knockdown of ErbB4 abrogated the effects of NRG1 on NF-κB, Bax levels and M2 microglial polarization. CONCLUSION: NRG1 inhibits the release of inflammatory factors in microglia and regulates the switching of the M1/M2 microglia phenotype, most likely via ErbB4-dependent inhibition of the NF-κB pathway.


Asunto(s)
Microglía , FN-kappa B , Neurregulina-1 , Neuroblastoma , Receptor ErbB-4 , Polaridad Celular/efectos de los fármacos , Humanos , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , FN-kappa B/metabolismo , Neurregulina-1/metabolismo , Neurregulina-1/farmacología , Neuroblastoma/metabolismo , Fenotipo , Receptor ErbB-4/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
19.
Int J Mol Sci ; 23(3)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35163566

RESUMEN

The Helicobacter pylori Neutrophil Activating Protein (HP-NAP) is endowed with immunomodulatory properties that make it a potential candidate for anticancer therapeutic applications. By activating cytotoxic Th1 responses, HP-NAP inhibits the growth of bladder cancer and enhances the anti-tumor activity of oncolytic viruses in the treatment of metastatic breast cancer and neuroendocrine tumors. The possibility that HP-NAP exerts its anti-tumor effect also by modulating the activity of innate immune cells has not yet been explored. Taking advantage of the zebrafish model, we examined the therapeutic efficacy of HP-NAP against metastatic human melanoma, limiting the observational window to 9 days post-fertilization, well before the maturation of the adaptive immunity. Human melanoma cells were xenotransplanted into zebrafish embryos and tracked in the presence or absence of HP-NAP. The behavior and phenotype of macrophages and the impact of their drug-induced depletion were analyzed exploiting macrophage-expressed transgenes. HP-NAP administration efficiently inhibited tumor growth and metastasis and this was accompanied by strong recruitment of macrophages with a pro-inflammatory profile at the tumor site. The depletion of macrophages almost completely abrogated the ability of HP-NAP to counteract tumor growth. Our findings highlight the pivotal role of activated macrophages in counteracting melanoma growth and support the notion that HP-NAP might become a new biological therapeutic agent for the treatment of metastatic melanomas.


Asunto(s)
Proteínas Bacterianas/administración & dosificación , Macrófagos/metabolismo , Melanoma/tratamiento farmacológico , Animales , Proteínas Bacterianas/inmunología , Línea Celular Tumoral , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Melanoma/inmunología , Metástasis de la Neoplasia , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
20.
Int J Mol Sci ; 23(3)2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35163619

RESUMEN

Classically activated M1 macrophages reprogram their metabolism towards enhanced glycolysis to obtain energy and produce pro-inflammatory cytokines after activation by mammalian target of rapamycin complex 1 (mTORC1) and hypoxia-inducible factor (HIF)-1α. Thus, a strategy that constrains M1 polarization of macrophages via downregulation of glycolysis is essential for treating chronic inflammatory diseases. Cassiae semen has pharmacological activity against various inflammatory diseases. However, it is unclear whether specific compounds within Cassia seeds affect M1 polarization of macrophages. Here, we investigated whether Cassiaside C napthopyrone from Cassiae semen inhibits M1 polarization by downregulating glycolysis. We found that Cassiaside C reduced expression of inducible nitric oxide synthase and cyclooxygenase-2 and the phosphorylation of nuclear factor kappa B, all of which are upregulated in lipopolysaccharide (LPS)/interferon (IFN)-γ-treated Raw264.7 cells and peritoneal macrophages. Moreover, Cassiaside C-treated macrophages showed marked suppression of LPS/IFN-γ-induced HIF-1α, pyruvate dehydrogenase kinase 1, and lactate dehydrogenase A expression, along with downregulation of the phosphoinositide 3-kinases (PI3K)/AKT/mTORC1 signaling pathway. Consequently, Cassiaside C attenuated enhanced glycolysis and lactate production, but rescued diminished oxidative phosphorylation, in M1 polarized macrophages. Thus, Cassiaside C dampens M1 polarization of macrophages by downregulating glycolysis, which could be exploited as a therapeutic strategy for chronic inflammatory conditions.


Asunto(s)
Polaridad Celular , Glucólisis , Glicósidos , Activación de Macrófagos , Macrófagos , Animales , Ratones , Regulación de la Expresión Génica , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7 , Transducción de Señal , Polaridad Celular/efectos de los fármacos , Glicósidos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA