Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.281
Filtrar
1.
J Immunol Res ; 2024: 7484490, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38455363

RESUMO

Macrophages are the immune cells of high-immunological plasticity, which can exert both pro- and anti-inflammatory activity, as well as repolarize their phenotype to the opposite or neutral one. In this regard, M2 macrophages of the tumor-associated stroma (TAS) are a promising therapeutic target in treating malignant neoplasms. Using FACS assay, we have estimated the CD11b+/Ly-6G+/Ly-6C+ fraction of macrophages from the peritoneum and TAS in intact healthy mice and those with developed Lewis carcinoma, both untreated and treated according to Karanahan technology in combination with group-specific macrophage activator (GcMAF-RF). As well, the pattern of pro- and anti-inflammatory cytokines mRNA expression in different groups of experimental and tumor-bearing animals was assessed. It was found that: (i) exposure of intact mice to GcMAF-RF results in the increased number of CD11b+/Ly-6C+ peritoneal macrophages and, at the same time, the expression pattern of cytokines in peritoneal macrophages switches from that characteristic of the mixed M1/M2 phenotype to that characteristic of the neutral M0 one; (ii) combination of Karanahan technology and GcMAF-RF treatment results in M0/M1 repolarization of TAS macrophages; (iii) in tumor-bearing mice, the response of peritoneal macrophages to such a treatment is associated with the induction of anti-inflammatory reaction, which is opposite to that in TAS macrophages.


Assuntos
Fatores Ativadores de Macrófagos , Macrófagos , Neoplasias , Proteína de Ligação a Vitamina D , Camundongos , Animais , Macrófagos Peritoneais/metabolismo , Citocinas/metabolismo , Neoplasias/patologia , Anti-Inflamatórios/metabolismo
2.
Int Immunopharmacol ; 128: 111554, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38262162

RESUMO

Macrophage is a vital factor in determining the fate of abdominal aortic aneurysm (AAA). The crosstalk between macrophage and other cells plays a crucial role in the development of aneurysm. Gasdermin D (GSDMD) is a vital executive protein of pyroptosis, which is a novel programmed cell death associated with inflammation. In this study, we identified aortic macrophage as the main expressing cell of GSDMD in AAA. Using Gsdmd-/-ApoE-/- mouse and AAV-F4/80-shGSDMD, we demonstrated the potential role of macrophage-derived GSDMD in AAA and aortic pyroptosis induced by Ang II in vivo. In vitro experiments showed that GSDMD promotes the pyroptosis of mouse primary peritoneal macrophages (MPMs), murine aortic vascular smooth muscle cells (MOVAS) and primary smooth muscle cells. Mechanistically, a mouse cytokine antibody array showed that Gsdmd-/- inhibited LPS + nigericin (LN)- induced secretion of multiple cytokines from MPMs. Furthermore, GSDMD is involved in the crosstalk between MPMs and MOVAS via cytokine secretion. This study provides a novel fundamental insight into macrophage-derived GSDMD in AAA and showed that GSDMD could be a promising therapeutic target for AAA.


Assuntos
Aneurisma da Aorta Abdominal , Piroptose , Animais , Camundongos , Angiotensina II/metabolismo , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Macrófagos Peritoneais/metabolismo , Miócitos de Músculo Liso/metabolismo
3.
Microbiol Spectr ; 12(1): e0347523, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38018982

RESUMO

IMPORTANCE: Sepsis is the consequence of a systemic bacterial infection that exacerbates the immune cell's activation via bacterial products, resulting in the augmented release of inflammatory mediators. A critical factor in the pathogenesis of sepsis is the primary component of the outer membrane of Gram-negative bacteria known as lipopolysaccharide (LPS), which is sensed by TLR4. For this reason, scientists have aimed to develop antagonists able to block TLR4 and, thereby the cytokine storm. We report here that a mixture of mu-class isoforms from the F. hepatica GST protein family administered intraperitoneally 1 h prior to a lethal LPS injection can modulate the dynamics and abundance of large peritoneal macrophages in the peritoneal cavity of septic mice while significantly suppressing the LPS-induced cytokine storm in a mouse model of septic shock. These results suggest that native F. hepatica glutathione S-transferase is a promising candidate for drug development against endotoxemia and other inflammatory diseases.


Assuntos
Fasciola hepatica , Sepse , Animais , Camundongos , Macrófagos Peritoneais/metabolismo , Lipopolissacarídeos/metabolismo , Fasciola hepatica/metabolismo , Escherichia coli/metabolismo , Síndrome da Liberação de Citocina/metabolismo , Receptor 4 Toll-Like/metabolismo , Macrófagos
4.
Nat Immunol ; 25(1): 155-165, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38102487

RESUMO

In mouse peritoneal and other serous cavities, the transcription factor GATA6 drives the identity of the major cavity resident population of macrophages, with a smaller subset of cavity-resident macrophages dependent on the transcription factor IRF4. Here we showed that GATA6+ macrophages in the human peritoneum were rare, regardless of age. Instead, more human peritoneal macrophages aligned with mouse CD206+ LYVE1+ cavity macrophages that represent a differentiation stage just preceding expression of GATA6. A low abundance of CD206+ macrophages was retained in C57BL/6J mice fed a high-fat diet and in wild-captured mice, suggesting that differences between serous cavity-resident macrophages in humans and mice were not environmental. IRF4-dependent mouse serous cavity macrophages aligned closely with human CD1c+CD14+CD64+ peritoneal cells, which, in turn, resembled human peritoneal CD1c+CD14-CD64- cDC2. Thus, major populations of serous cavity-resident mononuclear phagocytes in humans and mice shared common features, but the proportions of different macrophage differentiation stages greatly differ between the two species, and dendritic cell (DC2)-like cells were especially prominent in humans.


Assuntos
Macrófagos Peritoneais , Macrófagos , Humanos , Camundongos , Animais , Camundongos Endogâmicos C57BL , Macrófagos/metabolismo , Macrófagos Peritoneais/metabolismo , Diferenciação Celular , Células Dendríticas
5.
Biomed Khim ; 69(6): 394-402, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38153054

RESUMO

Hyperglycemia is one of the main damaging factors of diabetes mellitus (DM). The severity of this disease is most clearly manifested under conditions of the inflammatory process. In this work, we have studied the activation features of rat peritoneal macrophages (MPs) under conditions of high glucose concentration in vitro. Comparison of the independent and combined effects of streptozotocin-induced DM and hyperglycemia on proliferation and accumulation of nitrites in the MPs culture medium revealed similarity of their effects. Elevated glucose levels and, to a lesser extent, DM decreased basal proliferation and NO production by MPs in vitro. The use of the protein kinase C (PKC) activator, phorbol ester (PMA), abolished the proinflammatory effect of thrombin on PMs. This suggests the involvement of PKC in the effects of the protease. At the same time, the effect of thrombin on the level of nitrites in the culture medium demonstrates a pronounced dose-dependence, which was not recognized during evaluation of proliferation. Proinflammatory activation of MPs is potentiated by hyperglycemia, one of the main pathological factors of diabetes. Despite the fact that high concentrations of glucose have a significant effect on proliferation and NO production, no statistically significant differences were found between the responses of MPs obtained from healthy animals and from animals with streptozotocin-induced DM. This ratio was observed for all parameters studied in the work, during analysis of cell proliferation and measurement of nitrites in the culture medium. Thus, the results obtained indicate the leading role of elevated glucose levels in the regulation of MPs activation, which is comparable to the effect of DM and even "masks" it.


Assuntos
Diabetes Mellitus Experimental , Hiperglicemia , Ratos , Animais , Macrófagos Peritoneais/metabolismo , Nitritos , Estreptozocina/metabolismo , Estreptozocina/farmacologia , Trombina/metabolismo , Trombina/farmacologia , Hiperglicemia/metabolismo , Proteína Quinase C/metabolismo , Proteína Quinase C/farmacologia , Glucose/metabolismo
6.
Front Immunol ; 14: 1239592, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37965323

RESUMO

Persistent inflammation and associated pain significantly impact individuals' quality of life, posing substantial healthcare challenges. Proinflammatory cytokines, released by activated macrophages, play crucial roles in the development of chronic inflammatory conditions such as rheumatoid arthritis. To identify and evaluate potential therapeutic interventions targeting this process for mitigating inflammation and pain, we created myeloid cell-specific knockout of Vamp3 (vesicle-associated membrane protein 3) mice (Vamp3 Δmyel) by crossing LysM-Cre mice with newly engineered Vamp3flox/flox mice. Bone marrow-derived macrophages and peritoneal resident macrophages from Vamp3 Δmyel mice exhibited a significant reduction in TNF-α and IL-6 release compared to control mice. Moreover, Vamp3 deficiency led to decreased paw edema and ankle joint swelling induced by intraplantar injection of complete Freund's adjuvant (CFA). Furthermore, Vamp3 depletion also mitigated CFA-induced mechanical allodynia and thermal hyperalgesia. Mechanistically, Vamp3 loss ameliorated the infiltration of macrophages in peripheral sites of the hind paw and resulted in reduced levels of TNF-α and IL-6 in the CFA-injected paw and serum. RT-qPCR analysis demonstrated downregulation of various inflammation-associated genes, including TNF-α, IL-6, IL-1ß, CXCL11, TIMP-1, COX-2, CD68, and CD54 in the injected paw at the test day 14 following CFA administration. These findings highlight the novel role of Vamp3 in regulating inflammatory responses and suggest it as a potential therapeutic target for the development of novel Vamp-inactivating therapeutics, with potential applications in the management of inflammatory diseases.


Assuntos
Interleucina-6 , Fator de Necrose Tumoral alfa , Animais , Camundongos , Citocinas/metabolismo , Adjuvante de Freund , Hiperalgesia/induzido quimicamente , Hiperalgesia/genética , Inflamação/tratamento farmacológico , Macrófagos Peritoneais/metabolismo , Dor/induzido quimicamente , Qualidade de Vida , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteína 3 Associada à Membrana da Vesícula
7.
Biomolecules ; 13(11)2023 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-38002317

RESUMO

In this work, the incorporation of docosahexaenoic acid (DHA) in mouse resident peritoneal macrophages and its redistribution within the various phospholipid classes were investigated. Choline glycerophospholipids (PC) behaved as the major initial acceptors of DHA. Prolonged incubation with the fatty acid resulted in the transfer of DHA from PC to ethanolamine glycerophospholipids (PE), reflecting phospholipid remodeling. This process resulted in the cells containing similar amounts of DHA in PC and PE in the resting state. Mass spectrometry-based lipidomic analyses of phospholipid molecular species indicated a marked abundance of DHA in ether phospholipids. Stimulation of the macrophages with yeast-derived zymosan resulted in significant decreases in the levels of all DHA-containing PC and PI species; however, no PE or PS molecular species were found to decrease. In contrast, the levels of an unusual DHA-containing species, namely PI(20:4/22:6), which was barely present in resting cells, were found to markedly increase under zymosan stimulation. The levels of this phospholipid also significantly increased when the calcium-ionophore A23187 or platelet-activating factor were used instead of zymosan to stimulate the macrophages. The study of the route involved in the synthesis of PI(20:4/22:6) suggested that this species is produced through deacylation/reacylation reactions. These results define the increases in PI(20:4/22:6) as a novel lipid metabolic marker of mouse macrophage activation, and provide novel information to understand the regulation of phospholipid fatty acid turnover in activated macrophages.


Assuntos
Ácidos Docosa-Hexaenoicos , Macrófagos Peritoneais , Camundongos , Animais , Macrófagos Peritoneais/metabolismo , Zimosan , Fosfolipídeos/metabolismo , Ácidos Graxos/metabolismo
8.
PLoS One ; 18(10): e0291950, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37792774

RESUMO

System xc-, encoded by Slc7a11, is an antiporter responsible for exporting glutamate while importing cystine, which is essential for protein synthesis and the formation of thiol peptides, such as glutathione. Glutathione acts as a co-factor for enzymes responsible for scavenging reactive oxygen species. Upon exposure to bacterial products, macrophages exhibit a rapid upregulation of system xc-. This study investigates the impact of Slc7a11 deficiency on the functionality of peritoneal and bone marrow-derived macrophages. Our findings reveal that the absence of Slc7a11 results in significantly reduced glutathione levels, compromised mitochondrial flexibility, and hindered cytokine production in bone marrow-derived macrophages. Conversely, system xc- has a lesser impact on peritoneal macrophages in vivo. These results indicate that system xc- is essential for maintaining glutathione levels, mitochondrial functionality, and cytokine production, with a heightened importance under atmospheric oxygen tension.


Assuntos
Cistina , Ácido Glutâmico , Ácido Glutâmico/metabolismo , Cistina/metabolismo , Antiporters , Macrófagos Peritoneais/metabolismo , Glutationa/metabolismo , Citocinas/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo
9.
Int Immunopharmacol ; 120: 110297, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37207443

RESUMO

Overexpression of Staphylococcus aureus mediated CXCL8/CXCR1 axis is a major cause of sepsis and severe inflammatory diseases. This chemokine acts conjointly with various pro-inflammatory and anti-inflammatory cytokines that govern the severity of inflammation. The effects of different combinations of exogenous cytokines on CXCR1 expression in macrophages remain undetermined. Exogenous cytokine and anti-inflammatory cytokine therapy had been used to modulate CXCL8 and CXCR1 expression in peritoneal macrophages. Male Swiss albino mice were inoculated with live S. aureus (106 cells/ mouse) for the development of infection. Exogenous cytokines (TNF-α, IL-12, IFN-γ and IL-10) were administered intraperitoneally (single or combination) 24 h post S. aureus infection. The mice were sacrificed and peritoneal macrophages were isolated three days post infection. CXCL8, IL-12, IL-10 secretion, ROS generation and the bacterial phagocytic process had been evaluated. Western blot was used to study the expressions of TNFR1, IL-1R, CXCR1 and NF-κB. TNF-α, IL-12 and IFN-γ treatments aggravated CXCL8 and CXCR1 expression in the macrophages of infected mice. TNF-α + IFN-γ treatment was a major inducer of nitric oxide release and mediated maximum bacterial killing. IL-12 + TNF-α treatment was most potent in increasing ROS, CXCL8/CXCR1 expression through increased levels of TNFR1, IL-1R and NF-κB activation. IL-10 reversed the effects of exogenous cytokines but also impaired the bacterial clearance phenomenon in peritoneal lavage. Treatment with IL-12 + TNF-α + IL-10 was most effective in ameliorating oxidative stress, reduced CXCL8 release and expression levels of TNFR1, IL-1R, and NF-κB. Concludingly, IL-12 + TNF-α + IL-10 treatment mitigated CXCL8/CXCR1 expression and inflammatory signalling via downregulation of TNFR1-IL-1R-NF-κB pathway in peritoneal macrophages and inflammatory sequelae during S. aureus infection.


Assuntos
Receptores Tipo I de Fatores de Necrose Tumoral , Infecções Estafilocócicas , Animais , Masculino , Camundongos , Citocinas/metabolismo , Interleucina-10 , Interleucina-12/uso terapêutico , Macrófagos Peritoneais/metabolismo , NF-kappa B , Espécies Reativas de Oxigênio , Receptores de Interleucina-8A/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/uso terapêutico , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Fator de Necrose Tumoral alfa
10.
Adv Sci (Weinh) ; 10(11): e2206617, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36658699

RESUMO

Large peritoneal macrophages (LPMs) are long-lived, tissue-resident macrophages, formed during embryonic life, developmentally and functionally confined to the peritoneal cavity. LPMs provide the first line of defense against life-threatening pathologies of the peritoneal cavity, such as abdominal sepsis, peritoneal metastatic tumor growth, or peritoneal injuries caused by trauma, or abdominal surgery. Apart from their primary phagocytic function, reminiscent of primitive defense mechanisms sustained by coelomocytes in the coelomic cavity of invertebrates, LPMs fulfill an essential homeostatic function by achieving an efficient clearance of apoptotic, that is crucial for the maintenance of self-tolerance. Research performed over the last few years, in mice, has unveiled the mechanisms by which LPMs fulfill a crucial role in repairing peritoneal injuries and controlling microbial and parasitic infections, reflecting that the GATA6-driven LPM transcriptional program can be modulated by extracellular signals associated with pathological conditions. In contrast, recent experimental evidence supports that peritoneal tumors can subvert LPM metabolism and function, leading to the acquisition of a tumor-promoting potential. The remarkable functional plasticity of LPMs can be nevertheless exploited to revert tumor-induced LPM protumor potential, providing the basis for the development of novel immunotherapeutic approaches against peritoneal tumor metastasis based on macrophage reprogramming.


Assuntos
Macrófagos Peritoneais , Macrófagos , Animais , Camundongos , Macrófagos Peritoneais/metabolismo , Macrófagos/metabolismo , Homeostase
11.
J Hepatobiliary Pancreat Sci ; 30(3): 338-350, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35738898

RESUMO

BACKGROUND: Macrophages release large numbers of proinflammatory cytokines that trigger inflammatory cascade reactions, which promote the rapid development of severe acute pancreatitis (SAP) from local to systemic inflammation. The ability of mesenchymal stem cells (MSCs) to suppress inflammation is related to inhibition of M1 polarization of macrophages. Our previous studies revealed that caspase recruitment domain protein 9 (CARD9) was involved in SAP inflammation and activation of the CARD9-NF-κB signaling pathway plays an important proinflammatory role in SAP. At present, there is no effective treatment to control the inflammatory response in SAP. Therefore, the aim of the present study was to determine whether MSCs regulate the polarization of macrophages through the CARD9-NF-κB signaling pathway in SAP. METHODS: Short hairpin RNA interference technology and coculture in vitro were used to assess the activation status of the CARD9-NF-κB signal pathway in macrophages. Furthermore, flow cytometry was used to determine the polarization state of macrophages. RESULTS: The results showed MSCs inhibited CARD9 expression in vivo and in vitro (P < .05), alleviated inflammation induced by proinflammatory cytokines, and inhibited the phosphorylation of NF-κB in macrophages both in vivo and in vitro. Meanwhile, MSCs downregulated the CARD9-NF-κB signal pathway and inhibited M1 polarization of macrophages. CONCLUSION: In conclusion, MSCs regulate M1 polarization of peritoneal macrophages through the CARD9-NF-κB signaling pathway in SAP and transplantation of MSCs presents an effective treatment option for SAP.


Assuntos
Macrófagos Peritoneais , Células-Tronco Mesenquimais , NF-kappa B , Pancreatite , Humanos , Doença Aguda , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Citocinas/genética , Citocinas/metabolismo , Inflamação/genética , Inflamação/metabolismo , Macrófagos Peritoneais/metabolismo , Células-Tronco Mesenquimais/metabolismo , NF-kappa B/metabolismo , Pancreatite/genética , Pancreatite/metabolismo , Transdução de Sinais
12.
Mol Immunol ; 153: 10-24, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36402067

RESUMO

The JAK/STAT (Janus Kinase/Signal Transducer and Activator of Transcription) pathway plays a pivotal role in macrophage polarization, but other signaling routes may also be involved. The aim of this study was to reveal the relationship of activation between rat peritoneal macrophages and their polarization, to detect the signaling routes involved, and find selective protein kinase inhibitors decreasing the production of inflammatory proteins in activated peritoneal macrophages. Rat macrophages were elicited with i.p. casein injection. CD80 and CD206 markers, NOS2 (Nitric oxide synthase 2), arginase, cytokines and phagocytosis were investigated by ELISA (Enzyme Linked Immunosorbent Assay), Western Blot, fluorescent microscopic and flow cytometry. Statistical methods were ANOVA (Analysis Of Variance) and Student t-tests. Resident and elicited cells expressed both CD80 and CD206 polarization markers. The involvement of MAPK (mitogen-activated protein kinases) and JAK/STAT pathways in the polarization was evidenced by a phosphorylation array, supported by Western blotting, by cytokine markers and by the inhibitory effects of kinase inhibitors. The expression of NOS2 and inflammatory cytokines was higher in elicited cells suggesting their M1 polarization. This effect was reduced by the inhibitors of MAPK and JAK/STAT pathways. Phagocytosis was also higher in elicited macrophages and decreased by these inhibitors. Nevertheless, they cannot change macrophage polarization unambiguously, as levels of CD80 and CD206 markers were not changed. For comparison, human blood macrophages were also studied. Similar effects and several differences were observed between the two types of macrophages, suggesting the role of the previous differentiation in defining their characteristics. Selected anti-cancer protein kinase inhibitors of p38, MAPK and JAK/STAT pathways are possible candidates for the therapy of inflammatory diseases.


Assuntos
Citocinas , Macrófagos Peritoneais , Óxido Nítrico Sintase Tipo II , Inibidores de Proteínas Quinases , Animais , Humanos , Ratos , Citocinas/metabolismo , Janus Quinases , Macrófagos Peritoneais/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fagocitose , Inibidores de Proteínas Quinases/farmacologia
13.
Exp Gerontol ; 171: 112025, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36372284

RESUMO

Malnutrition is considered one of the most common problems in the elderly population worldwide and can significantly interfere in health evolution in these individuals, predisposing them to increased infection susceptibility. The immune response triggered by infections comprises several mechanisms, and macrophages play important roles in this response. This study aimed to evaluate mechanisms related to macrophage function in a model of protein malnutrition in the elderly. Two age groups (young: 3-5 months and elderly: 18-19 months) male C57BL/6NTac mice were subjected to protein malnutrition with a low-protein diet (2 %). The nutritional status, hemogram and number of peritoneal cells were affected by both age and nutritional status. Additionally, the spreading capacity as well as the phagocytic and fungicidal activity of peritoneal macrophages were affected by the nutritional status and age of the animal. Interestingly, the percentages of F4/80+/CD11b+ and CD86+ cells were reduced mostly in elderly animals, while the TLR-4+ population was more affected by nutritional status than by age. The production of pro-inflammatory cytokines such as TNF-α, IL-1α, and IL-6 was also influenced by nutritional status and/or by age, and malnourished animals of advanced age produced higher amounts of the anti-inflammatory cytokine IL-10. Furthermore, the phosphorylation ratio of the transcription factor NFκB (pNFκB/NFκB) was directly affected by the nutritional status, independently of age. Thus, these results allow us to conclude that aging and protein malnutrition compromise macrophage function, likely affecting their immune function, and in aged protein-malnourished animals, this impairment tends to be more pronounced.


Assuntos
Macrófagos Peritoneais , Desnutrição , Idoso , Humanos , Camundongos , Masculino , Animais , Macrófagos Peritoneais/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
14.
Front Immunol ; 13: 993788, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36263059

RESUMO

Changes in the function of peritoneal macrophages contribute to the homeostasis of the peritoneal immune microenvironment in endometriosis. The mechanism by which ectopic tissues escape phagocytic clearance by macrophages to achieve ectopic colonization and proliferation is unknown. The expression of CD163 in peritoneal macrophages in patients with endometriosis is increased, with the overexpression of MAPK, which can promote the M2-type polarization of macrophages and reduce their ability to phagocytose ectopic endometrial cells. As an upstream regulator of MAPK, MST1 expression is deficient in peritoneal macrophages of patients with endometriosis. This process is regulated by miR-887-5p, a noncoding RNA targeting MST1. Moreover, MST1-knockout macrophages secrete anti-inflammatory factor IL-10, which promotes autophagy of ectopic endometrial stromal cells. These results suggest that MST1 deficient macrophages may accelerate the autophagy of ectopic endometrium via IL-10 which was regulated by miR-887-5p.


Assuntos
Endometriose , MicroRNAs , Proteínas Serina-Treonina Quinases , Feminino , Humanos , Autofagia , Endometriose/metabolismo , Endometriose/patologia , Interleucina-10/metabolismo , Macrófagos Peritoneais/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Células Estromais/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
15.
Toxicon ; 217: 46-55, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-35981665

RESUMO

Crotoxin (CTX), the major toxin of Crotalus durissus terrificus snake venom, induces an inhibitory effect on tumor development and modulates the functions of macrophages (MØs), which play a key role as a defense mechanism against tumor growth. In early tumor progression stage, MØs are avidly phagocytic (inflammatory cell), releasing reactive nitrogen intermediates-RNI/ROI and cytokines TNF-α, IL-1ß, and IL-6. However, when the tumor has been developed, tumor-associated MØ (angiogenic cell) presents a decrease in the mentioned activities. We reported that CTX stimulates H2O2 release, NO production and secretion of cytokines by peritoneal MØs obtained from non-tumor-bearing rats. Considering that the mentioned mediators control tumor growth, it is mandatory to investigate whether CTX stimulates the production of these mediators by MØs obtained from tumor-bearing animals. The aim of this work was then to evaluate the CTX effect on metabolism and functions of peritoneal MØs obtained from Walker 256 tumor-bearing rats. For this purpose, male Wistar rats were subcutaneously inoculated in the right flank with 1 mL sterile suspension of 2 × 107 Walker 256 tumor cells. CTX (18 µg per animal) was subcutaneously administered in two protocols: a) on the 1st day of tumor cell injection and b) on the 4th day of tumor cell inoculation. In both protocols, MØs were obtaining on the 14th day of tumor cell inoculation to evaluate the release of H2O2, NO, and pro-inflammatory cytokines (IL-1ß, TNFα, and IL-6); maximal activity of hexokinase, glucose-6-phosphate dehydrogenase, citrate synthase, and 14CO2 production from [U-14C]-glucose and [U-14C]-glutamine. The treatment with CTX stimulated the release of NO, H2O2, and cytokines, and glucose and glutamine metabolism. Metabolic and functional changes induced by CTX were accompanied by a decrease of tumor growth as indicated by tumor fresh weight and diameter. These results indicate CTX not only as a scientific tool to investigate changes in metabolism and functions of peritoneal MØs but also for a better understanding of the mechanisms involved in tumor growth.


Assuntos
Crotoxina , Animais , Crotalus/metabolismo , Crotoxina/farmacologia , Citocinas/metabolismo , Glucose , Glutamina , Peróxido de Hidrogênio/metabolismo , Interleucina-6 , Macrófagos Peritoneais/metabolismo , Masculino , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa
16.
Cell Mol Biol (Noisy-le-grand) ; 68(4): 31-34, 2022 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-35988267

RESUMO

Ganoderma lucidum has been suggested as a natural immunomodulator. It is not yet clear exactly which combination of this extract is responsible for its immunomodulatory effects. Still, it appears that the 3-complement (CR3) receptor on the surface of immune cells acts as a receptor for beta-glucans (glucan-8), which is the main component of this extract. Since glucose-6-phosphate dehydrogenase (G6PD) plays a vital role in regulating macrophage functions, including nitric oxide production, we considered the effect of this extract on viability, G6PD enzyme activity, and nitric oxide (NO) production in peritoneal BALB/c macrophages. First, peritoneal macrophages of BALB/c mice were isolated and treated with concentrations (0.001, 0.01, 0.1, 1, 10, and 100 g/ml) of Ganoderma lucidum polysaccharide extract (GL-PS). After 24 hours of incubation by MTT test, we evaluated the viability of macrophages, and the effective dose was determined to be 0.1g/ml. To determine the specific activity of glucose-6-phosphates, they were incubated with GI-PS for 24 hours at a 0.1 mg/ml dose. Determination of protein concentration was obtained by the Bradford method in cell supernatant extract. Also, after 18 hours of incubation, the amount of nitric oxide (NO) production was measured using Grace colorimetric method. According to the results, a dose of 0.1µg/ml of Ganoderma lucidum polysaccharide extract had the most significant effect on the viability (stimulation coefficient) of peritoneal macrophages compared to other amounts (p <0.05). It was also found that a dose of 0.1µg/ml GL-PS increases NO production and the specific activity of the G6PD enzyme (p <0.05). Ganoderma lucidum, a medicinal fungus, is widely used in East Asian countries, especially in China, to increase the quality of life and longevity. After this study, we concluded that GL-PS extract has an immunomodulatory effect on macrophage activity. Therefore, the polysaccharide extract of this fungus can be used as a strengthening agent of the phagocytic system against infectious agents and pathogens such as the Leishmania parasite because of the production of nitric oxide by macrophages plays an essential role in defense against them.


Assuntos
Macrófagos Peritoneais , Reishi , Animais , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/metabolismo , Polissacarídeos/metabolismo , Polissacarídeos/farmacologia , Qualidade de Vida
17.
Drug Discov Ther ; 16(4): 148-153, 2022 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-36002309

RESUMO

Phenochalasin A, a unique phenol-containing cytochalasin produced by the marine-derived fungus Phomopsis sp. FT-0211, was originally discovered in a cell morphological assay of observing the inhibition of lipid droplet formation in mouse peritoneal macrophages. To investigate the mode of action and binding proteins, phenochalasin A was radio-labeled by 125I. Iodinated phenochalasin A exhibited the same biological activity as phenochalasin A. [125I]Phenochalasin A was found to be associated with an approximately 40 kDa protein, which was identified as G-actin. Furthermore, detail analyses of F-actin formation in Chinese hamster ovary cells (CHO-K1 cells) indicated that phenochalasin A (2 µM) caused elimination of F-actin formation on the apical site of the cells, suggesting that actin-oriented specific function(s) in cytoskeletal processes are affected by phenochalasin A.


Assuntos
Actinas , Gotículas Lipídicas , Actinas/análise , Actinas/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Citocalasinas/metabolismo , Citocalasinas/farmacologia , Indóis , Radioisótopos do Iodo , Lactonas , Gotículas Lipídicas/química , Gotículas Lipídicas/metabolismo , Macrófagos Peritoneais/química , Macrófagos Peritoneais/metabolismo , Camundongos , Fenóis
18.
Int Immunopharmacol ; 110: 109062, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35863257

RESUMO

The inflammatory focus is similar to the tumor microenvironment, which contains a complex milieu with immune cells and macrophages. The accumulation of cells promotes local pH and O2 tension decline (hypoxia). Local O2 tension decline activates hypoxia-inducible factor α and ß (HIF-1α and HIF-1ß adenosine triphosphate (ATP) release. ATP activates the P2X7 receptor and modulates ischemic/hypoxic conditions. Similarly, α1α may regulate P2X7 receptor expression in the hypoxic microenvironment. Therefore, we investigated P2X7 receptor function under simulated hypoxic conditions by pretreating peritoneal macrophages with mitochondrial electron transport chain complex inhibitors (simulated hypoxia). Treatment with mitochondrial electron transport chain complex inhibitors until three hours of exposure did not cause LDH release. Additionally, mitochondrial electron transport chain complex inhibitors increased ATP-induced P2X7 receptor function without being able to directly activate this receptor. Other P2 receptor subtypes do not appear to participate in this mechanism. Simulated hypoxia augmented HIF-1α levels and suppressed HIF-1α and P2X7 receptor antagonists. Similarly, simulated hypoxia increased ATP-induced dye uptake and inhibited HIF-1α antagonists. Another factor activated in simulated hypoxic conditions was the intracellular P2X7 receptor regulator PIP2. Treatment with HIF-1α agonists increased PIP2 levels and reversed the effects of HIF-1α and P2X7 receptor antagonists. Additionally, the improved ATP-induced dye uptake caused by the simulated hypoxia stimulus was inhibited by P2X7 receptor and PIP2 antagonists. Therefore, simulated hypoxia may augment P2X7 receptor activity for a pathway dependent on HIF-1α and PIP2 activation.


Assuntos
Macrófagos Peritoneais , Receptores Purinérgicos P2X7 , Trifosfato de Adenosina/metabolismo , Animais , Hipóxia Celular , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7/metabolismo
19.
Clin Exp Metastasis ; 39(5): 783-800, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35838814

RESUMO

In patients with pancreatic cancer (PC), the peritoneal cavity is the second-most common site of metastasis after the liver. Peritoneal macrophages (PMs) have been demonstrated to play a significant role in the peritoneal metastases of different cancers. Gemcitabine (GEM) is known to affect PC-associated immune cells, including macrophages. However, its effect on PMs and its possible clinical implication is yet to be investigated. In this study, mouse-derived PMs were treated with GEM ex vivo to analyze the polarization status. Production of GEM-induced reactive oxygen species (ROS) and reactive nitrogen species was evaluated using DCFH-DA, DAF-FM, and Griess assay. Antitumor effects of PMs on UN-KC-6141and UN-KPC-961 murine PC cells were evaluated in presence and absence of GEM in vitro. Similarly, effect of GEM on human THP-1 macrophage polarization and its tumoricidal effect was studied in vitro. Furthermore, the effect of GEM-treated PMs on peritoneal metastasis of UN-KC-6141 cells was evaluated in a syngeneic mouse model of PC. GEM upregulated M1 phenotype-associated molecular markers (Tnf-α and Inos) in vitro in PMs obtained from naïve mouse. Moreover, IL-4-induced M2-like PMs reverted to M1-like after GEM treatment. Co-culture of UN-KC-6141 and UN-KPC-961 cancer cells with PMs in the presence of GEM increased apoptosis of these cells, whereas cell death was markedly reduced after N-acetyl-L-cysteine treatment. Corroborating these findings co-culture of GEM-treated human THP-1 macrophages also induced cell death in MIAPaCa-2 cancer cells. GEM-treated PMs injected intraperitoneally along with UN-KC-6141 cells into mice extended survival period, but did not stop disease progression and mortality. Together, GEM induced M1-like polarization of PMs from naive and/or M2-polarized PMs in a ROS-dependent manner. GEM-induced M1-like PMs prompted cytotoxicity in PC cells and delayed disease progression in vivo.


Assuntos
Macrófagos Peritoneais , Macrófagos , Animais , Desoxicitidina/análogos & derivados , Progressão da Doença , Humanos , Macrófagos Peritoneais/metabolismo , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Gencitabina
20.
Nat Commun ; 13(1): 4406, 2022 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-35906202

RESUMO

Emerging evidence suggests that resident macrophages within tissues are enablers of tumor growth. However, a second population of resident macrophages surrounds all visceral organs within the cavities and nothing is known about these GATA6+ large peritoneal macrophages (GLPMs) despite their ability to invade injured visceral organs by sensing danger signals. Here, we show that GLPMs invade growing metastases that breach the visceral mesothelium of the liver via the "find me signal", ATP. Depleting GLPMs either by pharmacological or genetic tools, reduces metastases growth. Apoptotic bodies from tumor cells induces programmed cell death ligand 1 (PD-L1) upregulation on GLPMs which block CD8+ T cell function. Direct targeting of GLPMs by intraperitoneal but not intravenous administration of anti-PD-L1 reduces tumor growth. Thermal ablation of liver metastases recruits huge numbers of GLPMs and enables rapid regrowth of tumors. GLPMs contribute to metastatic growth and tumor recurrence.


Assuntos
Neoplasias Hepáticas , Macrófagos Peritoneais , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Fator de Transcrição GATA6/genética , Humanos , Macrófagos/metabolismo , Macrófagos Peritoneais/metabolismo , Recidiva Local de Neoplasia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...