Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

País/Región como asunto
País de afiliación
Intervalo de año de publicación
1.
An Acad Bras Cienc ; 96(2): e20231212, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38922279

RESUMEN

The tumor microenvironment (TME) harbors several cell types, such as tumor cells, immune cells, and non-immune cells. These cells communicate through several mechanisms, such as cell-cell contact, cytokines, chemokines, and extracellular vesicles (EVs). Tumor-derived vesicles are known to have the ability to modulate the immune response. Monocytes are a subset of circulating innate immune cells and play a crucial role in immune surveillance, being recruited to tissues where they differentiate into macrophages. In the context of tumors, it has been observed that tumor cells can attract monocytes to the TME and induce their differentiation into tumor-associated macrophages with a pro-tumor phenotype. Tumor-derived EVs have emerged as essential structures mediating this process. Through the transfer of specific molecules and signaling factors, tumor-derived EVs can shape the phenotype and function of monocytes, inducing the expression of cytokines and molecules by these cells, thus modulating the TME towards an immunosuppressive environment.


Asunto(s)
Vesículas Extracelulares , Macrófagos , Monocitos , Neoplasias , Microambiente Tumoral , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/fisiología , Humanos , Monocitos/inmunología , Microambiente Tumoral/inmunología , Neoplasias/inmunología , Neoplasias/patología , Macrófagos/inmunología , Citocinas , Diferenciación Celular
2.
Int J Mol Sci ; 21(24)2020 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-33352854

RESUMEN

Air pollution is mainly caused by burning of fossil fuels, such as diesel, and is associated with increased morbidity and mortality due to adverse health effects induced by inflammation and oxidative stress. Dimethyl fumarate (DMF) is a fumaric acid ester and acts as an antioxidant and anti-inflammatory agent. We investigated the potential therapeutic effects of DMF on pulmonary damage caused by chronic exposure to diesel exhaust particles (DEPs). Mice were challenged with DEPs (30 µg per mice) by intranasal instillation for 60 consecutive days. After the first 30 days, the animals were treated daily with 30 mg/kg of DMF by gavage for the remainder of the experimental period. We demonstrated a reduction in total inflammatory cell number in the bronchoalveolar lavage (BAL) of mice subjected to DEP + DMF as compared to those exposed to DEPs alone. Importantly, DMF treatment was able to reduce lung injury caused by DEP exposure. Intracellular total reactive oxygen species (ROS), peroxynitrite (OONO), and nitric oxide (NO) levels were significantly lower in the DEP + DMF than in the DEP group. In addition, DMF treatment reduced the protein expression of kelch-like ECH-associated protein 1 (Keap-1) in lung lysates from DEP-exposed mice, whereas total nuclear factor κB (NF-κB) p65 expression was decreased below baseline in the DEP + DMF group compared to both the control and DEP groups. Lastly, DMF markedly reduced DEP-induced expression of nitrotyrosine, glutathione peroxidase-1/2 (Gpx-1/2), and catalase in mouse lungs. In summary, DMF treatment effectively reduced lung injury, inflammation, and oxidative and nitrosative stress induced by chronic DEP exposure. Consequently, it may lead to new therapies to diminish lung injury caused by air pollutants.


Asunto(s)
Dimetilfumarato/farmacología , Estrés Oxidativo , Neumonía/etiología , Neumonía/metabolismo , Emisiones de Vehículos , Contaminantes Atmosféricos/efectos adversos , Animales , Biomarcadores , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones , FN-kappa B/metabolismo , Oxidación-Reducción , Neumonía/tratamiento farmacológico , Neumonía/patología , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Emisiones de Vehículos/toxicidad
3.
Helicobacter ; 24(3): e12573, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30907046

RESUMEN

BACKGROUND: Helicobacter pylori urease (HPU) is a key virulence factor that enables bacteria to colonize and survive in the stomach. We early demonstrated that HPU, independent of its catalytic activity, induced inflammatory and angiogenic responses in vivo and directly activated human neutrophils to produce reactive oxygen species (ROS). We have investigated the effects of HPU on endothelial cells, focusing on the signaling mechanism involved. METHODS: Monolayers of human microvascular endothelial cells (HMEC-1) were stimulated with HPU (up to 10 nmol/L): Paracellular permeability was accessed through dextran-FITC passage. NO and ROS production was evaluated using intracellular probes. Proteins or mRNA expressions were detected by Western blotting and fluorescence microscopy or qPCR assays, respectively. RESULTS: Treatment with HPU enhanced paracellular permeability of HMEC-1, preceded by VE-cadherin phosphorylation and its dissociation from cell-cell junctions. This caused profound alterations in actin cytoskeleton dynamics and focal adhesion kinase (FAK) phosphorylation. HPU triggered ROS and nitric oxide (NO) production by endothelial cells. Increased intracellular ROS resulted in nuclear factor kappa B (NF-κB) activation and upregulated expression of cyclooxygenase-2 (COX-2), hemeoxygenase-1 (HO-1), interleukin-1ß (IL-1ß), and intercellular adhesion molecule-1 (ICAM-1). Higher ICAM-1 and E-selectin expression was associated with increased neutrophil adhesion on HPU-stimulated HMEC monolayers. The effects of HPU on endothelial cells were dependent on ROS production and lipoxygenase pathway activation, being inhibited by esculetin. Additionally, HPU improved vascular endothelial growth factor receptor 2 (VEGFR-2) expression. CONCLUSION: The data suggest that the pro-inflammatory properties of HPU drive endothelial cell to a ROS-dependent program of differentiation that contributes to the progression of H pylori infection.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Infecciones por Helicobacter/inmunología , Helicobacter pylori/enzimología , Transducción de Señal/efectos de los fármacos , Ureasa/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Humanos , Inflamación , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Factores de Virulencia/farmacología
4.
Mem Inst Oswaldo Cruz ; 113(6): e140421, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29641644

RESUMEN

BACKGROUND Streptococcus agalactiae can causes sepsis, pneumonia, and meningitis in neonates, the elderly, and immunocompromised patients. Although the virulence properties of S. agalactiae have been partially elucidated, the molecular mechanisms related to reactive oxygen species (ROS) generation in infected human endothelial cells need further investigation. OBJECTIVES This study aimed to evaluate the influence of oxidative stress in human umbilical vein endothelial cells (HUVECs) during S. agalactiae infection. METHODS ROS production during S. agalactiae-HUVEC infection was detected using the probe CM-H2DCFDA. Microfilaments labelled with phalloidin-FITC and p47phox-Alexa 546 conjugated were analysed by immunofluorescence. mRNA levels of p47phox (NADPH oxidase subunit) were assessed using Real Time qRT-PCR. The adherence and intracellular viability of S. agalactiae in HUVECs with or without pre-treatment of DPI, apocynin (NADPH oxidase inhibitors), and LY294002 (PI3K inhibitor) were evaluated by penicillin/gentamicin exclusion. Phosphorylation of p47phox and Akt activation by S. agalactiae were evaluated by immunoblotting analysis. FINDINGS Data showed increased ROS production 15 min after HUVEC infection. Real-Time qRT-PCR and western blotting performed in HUVEC infected with S. agalactiae detected alterations in mRNA levels and activation of p47phox. Pre-treatment of endothelial cells with NADPH oxidase (DPI and apocynin) and PI3K/Akt pathway (LY294002) inhibitors reduced ROS production, bacterial intracellular viability, and generation of actin stress fibres in HUVECs infected with S. agalactiae. CONCLUSIONS ROS generation via the NADPH oxidase pathway contributes to invasion of S. agalactiae in human endothelial cells accompanied by cytoskeletal reorganisation through the PI3K/Akt pathway, which provides novel evidence for the involvement of oxidative stress in S. agalactiae pathogenesis.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , NADPH Oxidasas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Infecciones Estreptocócicas/metabolismo , Streptococcus agalactiae/metabolismo , Análisis de Varianza , Adhesión Bacteriana , Western Blotting , Células Endoteliales de la Vena Umbilical Humana/microbiología , Humanos , NADPH Oxidasas/análisis , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Valores de Referencia , Transducción de Señal/fisiología , Factores de Tiempo
5.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 139-151, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27677832

RESUMEN

Macrophage infiltration into adipose tissue (AT) is a hallmark of the chronic inflammatory response in obesity and is supported by an intense monocyte migration towards AT. Although it has been detected an increased proportion of circulating CD16+ monocyte subsets in obese subjects, the mechanisms underlying this effect and the contribution of these cells to the inflamed profile of obese AT are still poorly understood. We investigated whether factors secreted by human obese omental AT could polarize monocytes to CD16+ enriched phenotype, and how these changes could modify their migratory capacity towards adipose tissue itself. We show that explants of human obese omental AT, obtained during bariatric surgery, released higher levels of MIP1-α, TNFα, leptin and also VEGF, together with increasing amounts of microparticles (MP), when compared to explants of lean subcutaneous AT. A higher content of circulating MP derived from preadipocytes and leukocytes was also detected in plasma of obese subjects. Conditioned media or MP released from obese omental AT increased CD16 and CCR5 expression on CD14+CD16- monocytes and augmented their migratory capacity towards the conditioned media from obese omental AT, itself. This effect was inhibited when MIP1-α was neutralized. Additionally, we demonstrate that MP derived from obese omental AT carry and transfer TLR8 to monocytes, thus triggering an increase in CD16 expression in those cells. Our data shows a positive feedback loop between blood monocytes and obese omental AT, which releases chemotactic mediators and TLR8-enriched MP, thus inducing an up-regulation of CD16+ monocytes, favoring leukocyte infiltration in the obese omental AT.


Asunto(s)
Tejido Adiposo/inmunología , Micropartículas Derivadas de Células/inmunología , Monocitos/inmunología , Obesidad/inmunología , Receptores CCR5/inmunología , Receptores de IgG/inmunología , Receptor Toll-Like 8/inmunología , Tejido Adiposo/patología , Adulto , Micropartículas Derivadas de Células/patología , Femenino , Proteínas Ligadas a GPI/análisis , Proteínas Ligadas a GPI/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Masculino , Persona de Mediana Edad , Monocitos/patología , Obesidad/patología , Receptores CCR5/análisis , Receptores de IgG/análisis , Receptor Toll-Like 8/análisis
6.
Med Microbiol Immunol ; 204(6): 673-80, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25904542

RESUMEN

ExoU is a potent proinflammatory toxin produced by Pseudomonas aeruginosa, a major agent of severe lung infection and sepsis. Because inflammation is usually associated with oxidative stress, we investigated the effect of ExoU on free radical production and antioxidant defense mechanisms during the course of P. aeruginosa infection. In an experimental model of acute pneumonia, ExoU accounted for increased lipid peroxidation in mice lungs as soon as 3 h after intratracheal instillation of PA103 P. aeruginosa strain. The contribution of airway cells to the generation of a redox imbalance was assessed by in vitro tests carried out with A549 airway epithelial cells. Cultures infected with the ExoU-producing PA103 P. aeruginosa strain produced significantly increased concentrations of lipid hydroperoxides, 8-isoprostane, reactive oxygen intermediates, peroxynitrite and nitric oxide (NO), when compared to cells infected with exoU-deficient mutants. Overproduction of NO by PA103-infected cells likely resulted from overexpression of both inducible and endothelial NO synthase isoforms. PA103 infection was also associated with a significantly increased activity of superoxide dismutase (SOD) and decreased levels of reduced glutathione (GSH), a major antioxidant compound. Our findings unveil another potential mechanism of tissue damage during infection by ExoU-producing P. aeruginosa strains.


Asunto(s)
Proteínas Bacterianas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/microbiología , Pseudomonas aeruginosa/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Sepsis , Animales , Antioxidantes/metabolismo , Catalasa/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Femenino , Peroxidación de Lípido , Ratones , Superóxido Dismutasa/metabolismo
7.
Pharmacol Res ; 100: 73-84, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26253183

RESUMEN

Septic vascular dysfunction is characterized by hypotension and hyporeactivity to vasoconstrictors and nitric oxide (NO), reactive oxygen species and peroxynitrite have a prominent role in this condition. However, the mechanism whereby the vascular dysfunction is initiated is poorly understood. Based on previous studies of our group and the literature,we hypothesize that constitutive nitric oxide synthases (c-NOS) and peroxynitrite may play a role in the development of septic vascular dysfunction. Bacterial lipopolysaccharide (LPS) and interferon-γ (IFN) were used to stimulate rat aorta smooth muscle cells (A7r5) and rat aorta slices. This stimulation led to a rapid (within minutes) production of NO and superoxide anion, which led to peroxynitrite formation. When this rapid initial burst was reduced, through the inhibition of c-NOS and NADPH oxidases (NOX) or the scavenging of NO and superoxide the NF-κB activation, NOS-2 expression and nitrite production were significantly attenuated. Although vascular smooth muscle cells express both c-NOS isoforms, gene knockdown revealed that only NOS-1-dependent NO and peroxynitrite formation are important for the later NOS-2 expression. Similar findings were obtained by knockdown NOX-1 gene, one source of superoxide for peroxynitrite formation. Taking together, we show that smooth muscle cell activation by LPS/IFN leads to a rapid formation of NOS-1-derived NO and NOX-1-derived superoxide, forming peroxynitrite; and that this species act as a trigger for NOS-2 expression through NF-κB activation. Therefore, our findings suggest a critical role for NOS-1 and NOX-1 in the initiation of the vascular dysfunction associated with sepsis and septic shock.


Asunto(s)
Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Línea Celular , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , NADPH Oxidasas/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Choque Séptico/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Superóxidos/metabolismo
8.
Am J Physiol Cell Physiol ; 304(2): C170-9, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23114967

RESUMEN

In many gut chronic inflammatory conditions, intestinal epithelium (IE) is deprived of the protection of the mucus secreted by IE-specialized cells. In these events, bleeding and subsequent lysis of erythrocytes are common. This may lead to the release of high amounts of heme in the intestinal lumen, which interacts with IE. Previous works from our group have shown that heme itself is a proinflammatory molecule, activating a number of phlogistic signaling events in a nicotinamide adenine dinucleotide phosphate oxidase (NADPHox)-dependent manner. In this study, we aim to evaluate the effects of heme upon a well-established nontransformed small intestine epithelial cell lineage (IEC 6). Our results show that free heme evokes intracellular reactive oxygen species (ROS) production by IEC 6 cells, which is inhibited both by pharmacological inhibition with diphenyleneiodonium (10 µM), a NADPHox inhibitor, and small interfering RNA-mediated suppression of NOX1, a constitutive NADPHox isoform present in intestinal epithelial cells. Focal adhesion kinase phosphorylation and actin cytoskeleton polymerization are also induced by heme in a NADPHox-dependent manner. Heme increases monolayer permeability and redistributes key modulators of cell-cell adhesion as zona occludens-1 and E-cadherin proteins via NADPHox signaling. Heme promotes IEC 6 cell migration and proliferation, phenomena also regulated by NADPHox-derived ROS. Heme, in NADPHox-activating concentrations, is able to induce mRNA expression of IL-6, a cytokine implicated in inflammatory and tumorigenic responses. These data indicate a prominent role for heme-derived signaling in the pathophysiology of intestinal mucosa dysfunction and address an important role of NADPHox activity on the pathogenesis of intestinal inflammatory conditions.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Hemo/farmacología , Mucosa Intestinal/efectos de los fármacos , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Cadherinas/fisiología , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Duodeno/efectos de los fármacos , Duodeno/enzimología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Silenciador del Gen , Interleucina-6/biosíntesis , Mucosa Intestinal/enzimología , NADH NADPH Oxidorreductasas/antagonistas & inhibidores , NADH NADPH Oxidorreductasas/genética , NADPH Oxidasa 1 , NADPH Oxidasas/antagonistas & inhibidores , Compuestos Onio/farmacología , Permeabilidad/efectos de los fármacos , Fosforilación , Ratas , Transducción de Señal/genética , Proteína de la Zonula Occludens-1/fisiología
9.
Toxicon ; 223: 107011, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36584790

RESUMEN

Envenomation by Bothrops snakes and Apis mellifera bee may imply systemic disorders which affect well-perfused organs such as kidneys, a process that can lead to acute renal failure. Nevertheless, there is scarce information regarding a direct renal cell effect and the putative antagonism by antivenoms. Here the cytotoxic effect of B. jararacussu and A. mellifera venoms was evaluated in the renal proximal tubule cell line LLC-PK1, as well as the antagonism of this effect by heparin. B. jararacussu venom showed significant cytotoxicity as assessed by LDH release and MTT reduction, with a sharp decline of the cell number after 180 min (>90% at 50 µg/mL). A. mellifera venom produced a much faster and potent cytotoxic activity, conferring almost no viable cells after 15 min at 25 µg/mL. Phase contrast microscopy revealed that while B. jararacussu venom induced a progressive loss of cell adhesion and detachment, A. mellifera venom promoted a rapid plasma membrane disruption and nuclear condensation suggestive of necrotic cell death. Pre-incubation of both venoms with heparin for 30 min significantly reduced cytotoxicity. Our results demonstrate direct toxicity of B. jararacussu and A. mellifera venoms toward renal cells but with distinct kinetics and cell pattern, suggesting different mechanisms of action. In addition, the antagonistic, cytoprotective effect of heparin ascribes such compound as a promising drug for preventing renal failure from envenomation.


Asunto(s)
Antineoplásicos , Bothrops , Venenos de Crotálidos , Abejas , Animales , Heparina/farmacología , Antivenenos/farmacología , Venenos de Crotálidos/toxicidad , Riñón
10.
J Leukoc Biol ; 111(3): 585-596, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34043843

RESUMEN

Evidence shows that tumor cells abundantly produce and release extracellular vesicles (EVs) that can interact with stromal cells and modulate their functions. In the tumor neighborhood, neutrophils can assume both antitumor and pro-tumor phenotypes, known as TAN-N1 and TAN-N2, respectively. Nevertheless, the contribution of tumor-derived EVs to the modulation of TAN phenotypes is still poorly understood. The effects of EVs produced by a metastatic human melanoma cell line (MV3) on the differentiation and functional changes in human neutrophils were investigated. Treatment with MV3-derived EVs induced neutrophil chemotaxis through a signaling pathway involving the CXCR2/PI3K-Akt axis, prolonged neutrophil life span, promoted formation of neutrophil extracellular traps with poor elastase activity, and increased reactive oxygen species production. In contrast, EVs also increased the expression of TAN-N2 molecular markers (such as ARG1, CXCR4, and VEGF) in neutrophils. They also impaired oxide nitric and peroxynitrite production and diminished cytotoxic activity against melanoma cells, inducing neutrophils into a pro-tumor profile. Remarkably, EV-stimulated neutrophils did not exhibit phagocytic activity. These data suggested that melanoma-derived EVs could activate neutrophils, allowing their migration toward the tumor microenvironment, and driving these cells to a pro-tumor/N2 polarization, thus contributing to tumor progression.


Asunto(s)
Vesículas Extracelulares , Melanoma , Neutrófilos , Vesículas Extracelulares/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/patología , Neutrófilos/metabolismo , Neutrófilos/patología , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Microambiente Tumoral
11.
Cells ; 11(12)2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35741003

RESUMEN

Immune system cells, including neutrophils, are recruited by the tumor microenvironment as a site of chronic inflammation and begin to favor tumor growth. Neutrophils present in the tumor site are called tumor-associated neutrophils (TAN) and can present two phenotypes: N1 (antitumor) or N2 (pro-tumor). Evidence shows the high capacity of immune system cells to interact with extracellular vesicles (Evs) released by tumor cells. Evs can modulate the phenotype of cells within the immune system, contributing to tumor development. Here, we investigated the role of MDA-MB-231-derived Evs upon the polarization of neutrophils towards an N2 phenotype and the underlying mechanisms. We observed that neutrophils treated with Evs released by MDA cells (MDA-Evs) had their half-life increased, increased their chemotactic capacity, and released higher levels of NETs and ROS than neutrophils treated with non-tumoral Evs. We also observed that neutrophils treated with MDA-Evs released increased IL-8, VEGF, MMP9, and increased expression of CD184, an N2-neutrophil marker. Finally, neutrophils treated with MDA-Evs increased tumor cell viability. Our results show that MDA-Evs induce an N2-like phenotype, and the blockage of phosphatidylserine by annexin-V may be an essential agent counter-regulating this effect.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Supervivencia Celular , Vesículas Extracelulares/metabolismo , Humanos , Neoplasias/metabolismo , Neutrófilos/metabolismo , Microambiente Tumoral
12.
Cancers (Basel) ; 13(13)2021 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-34283044

RESUMEN

Extracellular vesicles (EVs) are crucial elements that sustain the communication between tumor cells and their microenvironment, and have emerged as a widespread mechanism of tumor formation and metastasis. In obesity, the adipose tissue becomes hypertrophic and hyperplastic, triggering increased production of pro-inflammatory adipokines, such as tumor necrosis factor α, interleukin 6, interleukin 1, and leptin. Furthermore, obese adipose tissue undergoes dysregulation in the cargo content of the released EVs, resulting in an increased content of pro-inflammatory proteins, fatty acids, and oncogenic microRNAs. These alterations drive obesity-associated inflammatory responses both locally and systemically. After being ignored for a long time, adipose tissues have recently received considerable attention as a major player in tumor microenvironment-linked obesity and cancer. The role of adipose tissue in the establishment and progression of cancer is reinforced by its high plasticity and inflammatory content. Such a relationship may be established by direct contact between adipocytes and cancer cells within the microenvironment or systemically, via EV-mediated cell-to-cell communication. Here, we highlight cues evidencing the influence of adipose tissue-derived EVs on the hallmarks of cancer, which are critical for tumor malignancy.

13.
Metabolites ; 11(12)2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34940560

RESUMEN

Acute mesenteric ischemia, caused by an abrupt interruption of blood flow in the mesenteric vessels, is associated with high mortality. When treated with surgical interventions or drugs to re-open the vascular lumen, the reperfusion process itself can inflict damage to the intestinal wall. Ischemia and reperfusion injury comprise complex mechanisms involving disarrangement of the splanchnic microcirculatory flow and impairment of the mitochondrial respiratory chain due to initial hypoxemia and subsequent oxidative stress during the reperfusion phase. This pathophysiologic process results in the production of large amounts of reactive oxygen (ROS) and nitrogen (RNS) species, which damage deoxyribonucleic acid, protein, lipids, and carbohydrates by autophagy, mitoptosis, necrosis, necroptosis, and apoptosis. Fluorescence-based systems using molecular probes have emerged as highly effective tools to monitor the concentrations and locations of these often short-lived ROS and RNS. The timely and accurate detection of both ROS and RNS by such an approach would help to identify early injury events associated with ischemia and reperfusion and increase overall clinical diagnostic sensitivity. This abstract describes the pathophysiology of intestinal ischemia and reperfusion and the early biological laboratory diagnosis using fluorescent molecular probes anticipating clinical decisions in the face of an extremely morbid disease.

14.
Toxins (Basel) ; 13(12)2021 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-34941745

RESUMEN

The significant incidence of deforestation in South America culminates in the contact of humans with typical forests species. Among these species, one may highlight Lonomia obliqua caterpillar, which, when touched by humans, can poison them through their bristles. Therefore, better acknowledging the mechanisms involved in envenomation caused by Lonomia obliqua caterpillar bristle extract (LOCBE) may contribute to further treatments. Recently, we demonstrated that LOCBE induces a pro-inflammatory profile in endothelial cells; thus, we decided to investigate the effects of LOCBE on human polymorphonuclear neutrophils (PMN), which are the first leukocytes that migrate to the inflammatory focus. Our results showed that treatment with LOCBE induced PMN chemotaxis together with alterations in actin cytoskeleton and focal adhesion kinase (FAK) activation, favoring migration. Concurrently, LOCBE induced PMN adhesion to matrix proteins, such as collagen IV, fibronectin, and fibrinogen. Moreover, we observed that LOCBE attenuated PMN apoptosis and increased reactive oxygen species (ROS) production together with nuclear factor kB (NF-κB) activation-a redox-sensitive transcription factor-as well as interleukin (IL)-1ß and IL-8 release. We call attention to the ROS-dependent effect of LOCBE on increased cell migration once an antioxidant treatment reverted it. In summary, we report that LOCBE activates PMN, inducing pro-inflammatory responses modulated by ROS.


Asunto(s)
Venenos de Artrópodos/toxicidad , Lepidópteros/fisiología , Neutrófilos/efectos de los fármacos , Animales , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Quimiotaxis , Cricetinae , Humanos , Integumento Común , Larva/fisiología , Especies Reactivas de Oxígeno/metabolismo , Piel/efectos de los fármacos
15.
Toxicol Lett ; 341: 11-22, 2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-33472085

RESUMEN

AIMS: Accidental contact with the Lonomia obliqua caterpillar is a common event in southern Brazil. Envenomed victims present consumption coagulopathy, which can evolve to acute kidney injury (AKI). In the present study, we searched for AKI biomarkers and changes in molecular pathway signatures through urine proteomic analysis. METHODOLOGY: Male Wistar rats were injected with L. obliqua venom (1.5 mg/kg, via s.c.) or 0.9 % NaCl and distributed into metabolic cages. After 24 h, urine was obtained, and the set of differentially regulated proteins was analyzed by MudPIT technology in an OrbiTRAP mass spectrometer. RESULTS: L. obliqua venom leads to an increase in urine output and water and electrolyte excretion and to an increase in the albumin to creatine ratio in urine. The proteomic analysis revealed an up-regulation of tubular injury biomarkers, such as neutrophil-gelatinase associated lipocalin (NGAL) and cystatin C, in urine from envenomed rats. Several components related to the heme scavenging system were up-regulated or exclusively identified in urine from envenomed animals. There was an increase in urinary heme levels and hemoglobin subunits, hemopexin, haptoglobin, and biliverdin reductase. Similarly, kinin- and angiotensin-generating/degrading peptidases, such as kallikreins, neprilysin, plasmin, dipeptidyl peptidase IV, cathepsin D, kininogen, and neutral, basic, glutamyl, and acidic aminopeptidases, were also up-regulated in urine. CONCLUSIONS: L. obliqua envenomation induced tubular and glomerular injury, probably involving heme/hemoglobin toxicity and an imbalance in the kinin/angiotensin generating/degrading system.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Aminopeptidasas/metabolismo , Venenos de Artrópodos/toxicidad , Hemoglobinuria , Lepidópteros , Proteómica , Aminopeptidasas/química , Animales , Hemo , Hemoglobinas , Larva/fisiología , Masculino , Ratas , Ratas Wistar , Urinálisis , Orina/química
16.
Toxins (Basel) ; 13(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33466223

RESUMEN

Massive, Africanized honeybee attacks have increased in Brazil over the years. Humans and animals present local and systemic effects after envenomation, and there is no specific treatment for this potentially lethal event. This study evaluated the ability of a new Apilic antivenom, which is composed of F(ab')2 fraction of specific immunoglobulins in heterologous and hyperimmune equine serum, to neutralize A. mellifera venom and melittin, in vitro and in vivo, in mice. Animal experiments were performed in according with local ethics committee license (UFRJ protocol no. DFBCICB072-04/16). Venom dose-dependent lethality was diminished with 0.25-0.5 µL of intravenous Apilic antivenom/µg honeybee venom. In vivo injection of 0.1-1 µg/g bee venom induced myotoxicity, hemoconcentration, paw edema, and increase of vascular permeability which were antagonized by Apilic antivenom. Cytotoxicity, assessed in renal LLC-PK1 cells and challenged with 10 µg/mL honeybee venom or melittin, was neutralized by preincubation with Apilic antivenom, as well the hemolytic activity. Apilic antivenom inhibited phospholipase and hyaluronidase enzymatic activities. In flow cytometry experiments, Apilic antivenom neutralized reduction of cell viability due to necrosis by honeybee venom or melittin. These results showed that this antivenom is effective inhibitor of honeybee venom actions. Thus, this next generation of Apilic antivenom emerges as a new promising immunobiological product for the treatment of massive, Africanized honeybee attacks.


Asunto(s)
Antivenenos/uso terapéutico , Venenos de Abeja/antagonistas & inhibidores , Mordeduras y Picaduras/tratamiento farmacológico , Meliteno/antagonistas & inhibidores , Animales , Anticuerpos/sangre , Abejas , Brasil , Línea Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Hemólisis/efectos de los fármacos , Caballos , Hialuronoglucosaminidasa/antagonistas & inhibidores , Fragmentos Fab de Inmunoglobulinas/uso terapéutico , Inyecciones Intradérmicas , Células LLC-PK1 , Dosificación Letal Mediana , Masculino , Ratones , Modelos Animales , Pruebas de Neutralización , Fosfolipasas/antagonistas & inhibidores , Porcinos
17.
Sci Rep ; 11(1): 15833, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-34349175

RESUMEN

Metastatic disease remains the leading cause of death in cancer and understanding the mechanisms involved in tumor progression continues to be challenging. This work investigates the role of manganese in tumor progression in an in vivo model of tumor growth. Our data revealed that manganese accumulates within primary tumors and secondary organs as manganese-rich niches. Consequences of such phenomenon were investigated, and we verified that short-term changes in manganese alter cell surface molecules syndecan-1 and ß1-integrin, enhance collective cell migration and invasive behavior. Long-term increased levels of manganese do not affect cell growth and viability but enhance cell migration. We also observed that manganese is secreted from tumor cells in extracellular vesicles, rather than in soluble form. Finally, we describe exogenous glycosaminoglycans that counteract manganese effects on tumor cell behavior. In conclusion, our analyses describe manganese as a central element in tumor progression by accumulating in Mn-rich niches in vivo, as well as in vitro, affecting migration and extracellular vesicle secretion in vitro. Manganese accumulation in specific regions of the organism may not be a common ground for all cancers, nevertheless, it represents a new aspect of tumor progression that deserves special attention.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Pulmonar de Lewis/patología , Movimiento Celular , Manganeso/metabolismo , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nutrients ; 12(9)2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32947825

RESUMEN

In obesity, the dysfunctional adipose tissue (AT) releases increased levels of proinflammatory adipokines such as TNFα, IL-6, and IL-1ß and free fatty acids (FFAs), characterizing a chronic, low-grade inflammation. Whilst FFAs and proinflammatory adipokines are known to elicit an inflammatory response within AT, their relative influence upon preadipocytes, the precursors of mature adipocytes, is yet to be determined. Our results demonstrated that the conditioned medium (CM) derived from obese AT was rich in FFAs, which guided us to evaluate the role of TLR4 in the induction of inflammation in preadipocytes. We observed that CM derived from obese AT increased reactive oxygen species (ROS) levels and NF-ĸB nuclear translocation together with IL-6, TNFα, and IL-1ß in 3T3-L1 cells in a TLR4-dependent manner. Furthermore, TLR4 signaling was involved in the increased expression of C/EBPα together with the release of leptin, adiponectin, and proinflammatory mediators, in response to the CM derived from obese AT. Our results suggest that obese AT milieu secretes lipokines, which act in a combined paracrine/autocrine manner, inducing inflammation in preadipocytes via TLR4 and ROS, thus creating a paracrine loop that facilitates the differentiation of adipocytes with a proinflammatory profile.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Inflamación/metabolismo , Obesidad/metabolismo , Receptor Toll-Like 4/metabolismo , Adipoquinas/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Transducción de Señal
19.
J Innate Immun ; 11(4): 330-346, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30557873

RESUMEN

Exposition of neutrophils (polymorphonuclear neutrophils, PMNs) to bacterial products triggers exacerbated activation of these cells, increasing their harmful effects on host tissues. We evaluated the possibility of interfering with the classic immune innate responses of human PMNs exposed to bacterial endotoxin (lipopolysaccharide, LPS), and further stimulated with bacterial formyl peptide (N-formyl-methionine-leucine-phenylalanine, fMLP). We showed that the low- molecular-weight fucoidan (LMW-Fuc), a polysaccharide extracted from brown algae, attenuated the exacerbated activation induced by fMLP on LPS-primed PMNs, in vitro, impairing chemotaxis, NET formation, and the pro-survival and pro-oxidative effects. LMW-Fuc also inhibited the activation of canonical signaling pathways, AKT, bad, p47phox and MLC, activated by the exposition of PMN to bacterial products. The activation of PMN by sequential exposure to LPS and fMLP induced the release of L-selectin+ microparticles, which were able to trigger extracellular reactive oxygen species production by fresh PMNs and macrophages. Furthermore, we observed that LMW-Fuc inhibited microparticle release from activated PMN. In vivo experiments showed that circulating PMN-derived microparticles could be detected in mice exposed to bacterial products (LPS/fMLP), being downregulated in animals treated with LMW-Fuc. The data highlight the autocrine and paracrine role of pro-inflammatory microparticles derived from activated PMN and demonstrate the anti-inflammatory effects of LMW-Fuc on these cells.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Trampas Extracelulares/metabolismo , Selectina L/metabolismo , Neutrófilos/inmunología , Polisacáridos/farmacología , Animales , Supervivencia Celular , Células Cultivadas , Quimiotaxis , Humanos , Inmunidad Innata , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos BALB C , N-Formilmetionina Leucil-Fenilalanina/inmunología , Activación Neutrófila , Estrés Oxidativo , Phaeophyceae/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
20.
PLoS Negl Trop Dis ; 13(2): e0007197, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30763408

RESUMEN

BACKGROUND: Lonomia obliqua venom is nephrotoxic and acute kidney injury (AKI) is the main cause of death among envenomed victims. Mechanism underlying L. obliqua-induced AKI involves renal hypoperfusion, inflammation, tubular necrosis and loss of glomerular filtration and tubular reabsorption capacities. In the present study, we aimed to investigate the contribution of kallikrein to the hemodynamic instability, inflammation and consequent renal and vascular impairment. METHODOLOGY/PRINCIPAL FINDINGS: Addition of L. obliqua venom to purified prekallikrein and human plasma in vitro or to vascular smooth muscle cells (VSMC) in culture, was able to generate kallikrein in a dose-dependent manner. Injected in rats, the venom induced AKI and increased kallikrein levels in plasma and kidney. Kallikrein inhibition by aprotinin prevented glomerular injury and the decrease in glomerular filtration rate, restoring fluid and electrolyte homeostasis. The mechanism underlying these effects was associated to lowering renal inflammation, with decrease in pro-inflammatory cytokines and matrix metalloproteinase expression, reduced tubular degeneration, and protection against oxidative stress. Supporting the key role of kallikrein, we demonstrated that aprotinin inhibited effects directly associated with vascular injury, such as the generation of intracellular reactive oxygen species (ROS) and migration of VSMC induced by L. obliqua venom or by diluted plasma obtained from envenomed rats. In addition, kallikrein inhibition also ameliorated venom-induced blood incoagulability and decreased kidney tissue factor expression. CONCLUSIONS/SIGNIFICANCE: These data indicated that kallikrein and consequently kinin release have a key role in kidney injury and vascular remodeling. Thus, blocking kallikrein may be a therapeutic alternative to control the progression of venom-induced AKI and vascular disturbances.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Venenos de Artrópodos/toxicidad , Calicreínas/antagonistas & inhibidores , Mariposas Nocturnas/fisiología , Lesión Renal Aguda/prevención & control , Animales , Aprotinina , Trastornos de la Coagulación Sanguínea/inducido químicamente , Modelos Animales de Enfermedad , Tasa de Filtración Glomerular , Larva/fisiología , Masculino , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA