Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros













Intervalo de año de publicación
1.
Cells ; 12(4)2023 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-36831188

RESUMEN

Obesity is an alarming disease that favors the upset of other illnesses and enhances mortality. It is spreading fast worldwide may affect more than 1 billion people by 2030. The imbalance between excessive food ingestion and less energy expenditure leads to pathological adipose tissue expansion, characterized by increased production of proinflammatory mediators with harmful interferences in the whole organism. Bone tissue is one of those target tissues in obesity. Bone is a mineralized connective tissue that is constantly renewed to maintain its mechanical properties. Osteoblasts are responsible for extracellular matrix synthesis, while osteoclasts resorb damaged bone, and the osteocytes have a regulatory role in this process, releasing growth factors and other proteins. A balanced activity among these actors is necessary for healthy bone remodeling. In obesity, several mechanisms may trigger incorrect remodeling, increasing bone resorption to the detriment of bone formation rates. Thus, excessive weight gain may represent higher bone fragility and fracture risk. This review highlights recent insights on the central mechanisms related to obesity-associated abnormal bone. Publications from the last ten years have shown that the main molecular mechanisms associated with obesity and bone loss involve: proinflammatory adipokines and osteokines production, oxidative stress, non-coding RNA interference, insulin resistance, and changes in gut microbiota. The data collection unveils new targets for prevention and putative therapeutic tools against unbalancing bone metabolism during obesity.


Asunto(s)
Resorción Ósea , Osteoclastos , Humanos , Osteoclastos/metabolismo , Osteoblastos/metabolismo , Huesos , Resorción Ósea/metabolismo , Obesidad/metabolismo
2.
Toxicon ; 217: 87-95, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-35981667

RESUMEN

Integrins are transmembrane heterodimeric glycoproteins, present in most cell types that act as mechanoreceptors, connecting extracellular matrix proteins to the cytoskeleton of the cell, mediating several physiological and pathological processes. The disintegrins are peptides capable of modulating the activity of integrins, such as αIIbß3, responsible for the platelet aggregation and αvß3, related to angiogenesis. The aim of this study was to produce the recombinant disintegrin jarastatin (rJast), to evaluate its secondary structure and biological activity. rJast was expressed in the yeast Komagataella phaffii (earlier Pichia pastoris) purified using molecular exclusion chromatography and the internal sequence and molecular mass were confirmed by mass spectrometry. The yield was approximately 40 mg/L of culture. rJast inhibited platelet aggregation induced by 2-4 µM ADP, 10 nM thrombin, and 1 µg/mL collagen (IC50 of 244.8 nM, 166.3 nM and 223.5 nM, respectively). It also blocked the adhesion of platelets to collagen under continuous flow in approximately 60% when used 1 µM. We also evaluated the effect of rJast on HMEC-1 cells. rJast significantly inhibited the adhesion of these cells to vitronectin, as well as cell migration (IC50 1.77 µM) without changing the viability. Conclusions: rJast was successfully expressed with activity in human platelets aggregation identical to the native molecule. Also, rJast inhibits adhesion and migration of endothelial cells. Thus, being relevant for the development of anti-thrombotic and anti-angiogenic drugs.


Asunto(s)
Venenos de Crotálidos , Desintegrinas , Adhesión Celular , Movimiento Celular , Colágeno , Venenos de Crotálidos/química , Desintegrinas/química , Células Endoteliales , Humanos , Integrinas , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/química , Inhibidores de Agregación Plaquetaria/farmacología
3.
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
5.
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
6.
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
7.
Nanotechnology ; 32(43)2021 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-34271563

RESUMEN

The urgency for new materials in oncology is immediate. In this study we have developed the g-C3N4, a graphitic-like structure formed by periodically linked tris-s-triazine units. The g-C3N4has been synthesized by a simple and fast thermal process. XRD has shown the formation of the crystalline sheet with a compacted structure. The graphite-like structure and the functional groups have been shown by Raman and FTIR spectroscopy. TEM image and AFM revealed the porous composed of five or six C-N layers stacked. DRS and Photoluminescence analyses confirmed the structure with band gap of 2.87 eV and emission band at 448 nm in different wavelengths excitation conditions. The biological results showed inhibitory effect on cancer cell lines and non-toxic effect in normal cell lines. To the best of our knowledge, this is the first work demonstrating the cytotoxic effects of 2D g-C3N4in a cancer cell line, without any external or synergistic influence. The biodistribution/tissue accumulation showed that g-C3N4present a tendency to accumulation on the lung in the first 2 h, but after 24 h the profile of the biodistribution change and it is found mainly in the liver. Thus, 2D-g-C3N4showed great potential for the treatment of several cancer types.


Asunto(s)
Supervivencia Celular , Grafito/síntesis química , Grafito/metabolismo , Compuestos de Nitrógeno/síntesis química , Compuestos de Nitrógeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Humanos , Distribución Tisular
8.
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.

9.
J Mol Endocrinol ; 67(1): 15-26, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34045365

RESUMEN

In obesity, high levels of TNF-α in the bone marrow microenvironment induce the bone marrow-mesenchymal stem cells (BM-MSCs) towards a pro-adipogenic phenotype. Here, we investigated the effect of obesity on the migratory potential of BM-MSCs and their fate towards the adipose tissues. BM-MSCs were isolated from male C57Bl/06 mice with high-fat diet-induced obesity. The migratory potential of the BM-MSCs, their presence in the subcutaneous (SAT) and the visceral adipose tissues (VAT), and the possible mechanisms involved were investigated. Obesity did not affect MSC content in the bone marrow but increased the frequency of MSCs in blood, SAT, and VAT. In these animals, the SAT adipocytes presented a larger area, without any changes in adipokine production or the Sdf-1α gene expression. In contrast, in VAT, obesity increased leptin and IL-10 levels but did not modify the size of the adipocytes. The BM-MSCs from obese animals presented increased spontaneous migratory activity. Despite the augmented expression of Cxcr4, these cells exhibited decreased migratory response towards SDF-1α, compared to that of BM-MSCs from lean mice. The PI3K-AKT pathway activation seems to mediate the migration of BM-MSCs from lean mice, but not from obese mice. Additionally, we observed an increase in the spontaneous migration of BM-MSCs from lean mice when they were co-cultured with BM-HCs from obese animals, suggesting a paracrine effect. We concluded that obesity increased the migratory potential of the BM-MSCs and induced their accumulation in VAT, which may represent an adaptive mechanism in response to chronic nutrient overload.


Asunto(s)
Grasa Intraabdominal/patología , Células Madre Mesenquimatosas/patología , Obesidad/patología , Animales , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Grasa Intraabdominal/efectos de los fármacos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Obesos , Comunicación Paracrina/efectos de los fármacos , Receptores CXCR4/metabolismo , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Grasa Subcutánea/efectos de los fármacos , Grasa Subcutánea/patología , Factor de Necrosis Tumoral alfa/farmacología
10.
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
11.
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
12.
Endocr Relat Cancer ; 27(10): 571-582, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32738129

RESUMEN

Obesity is a chronic low-grade inflammatory condition that strongly impacts breast cancer. Aside from inflammatory mediators, obese adipose tissue (AT) secretes high amounts of extracellular vesicles (EVs), which are capable of transferring molecules to target cells and promoting cell-to-cell communication. Here, we investigated how soluble mediators and EVs secreted by human obese AT influence MCF-7 and MDA-MB-231 mammary adenocarcinoma cell lines by modulating cell proliferation, migration, invasion, and signaling pathways. Both cell lineages were stimulated with conditioned media (CM) or EVs obtained from cultures of AT explants collected from lean or obese individuals who underwent plastic or bariatric surgeries, respectively. EVs derived from obese AT increased the proliferative potential of both cell lines and further potentiated the migratory and invasive properties of MDA-MB-231 cells. The proliferative effects of CM and EVs on MCF-7 cells were dependent on ERK/MAPK pathway activation, while the migration and invasiveness of MDA-MB-231 cells were dependent on PI3K/AKT pathway activation. Furthermore, CM derived from obese AT potentiated the pro-angiogenic effect of MDA-MB-231 on endothelial cells. We also detected that EVs derived from obese AT were enriched in leptin and bioactive matrix metallopeptidase 9 (MMP9), and stimulation of MDA-MD-231 cells with those EVs or CM derived from obese AT potentiated the release of MMP9 by those cells. Our data indicate that obese AT secretes molecules and EVs with pro-tumoral activities capable of increasing breast cancer cell malignancy and provide strong evidence of the key role of AT-derived EV signaling in the tumor microenvironment.


Asunto(s)
Tejido Adiposo/fisiopatología , Neoplasias de la Mama/fisiopatología , Vesículas Extracelulares/metabolismo , Obesidad/complicaciones , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Ratones , Obesidad/patología
13.
Int J Mol Sci ; 21(8)2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-32340328

RESUMEN

Alterations in the composition and architecture of the extracellular matrix (ECM) can influence cancer growth and dissemination. During epithelial-mesenchymal transition (EMT), epithelial cells assume a mesenchymal cell phenotype, changing their adhesion profiles from cell-cell contacts to cell-matrix interactions, contributing to metastasis. Breast cancer cells present at different stages of differentiation, producing distinct ECMs in the same tumor mass. However, the contribution of ECM derived from metastatic tumor cells to EMT is unclear. Here, we showed the mechanisms involved in the interaction of MCF-7, a low-metastatic, epithelial breast cancer cell line, with the ECM produced by a high metastatic breast tumor cell, MDA-MB-231 (MDA-ECM). MDA-ECM induced morphological changes in MCF-7 cells, decreased the levels of E-cadherin, up-regulated mesenchymal markers, and augmented cell migration. These changes were accompanied by the activation of integrin-associated signaling, with increased phosphorylation of FAK, ERK, and AKT and activation canonical TGF-ß receptor signaling, enhancing phosphorylation of SMAD2 and SMAD4 nuclear translocation in MCF-7 cells. Treatment with Kistrin (Kr), a specific ligand of integrin αvß3 EMT induced by MDA-ECM, inhibited TGF-ß receptor signaling in treated MCF-7 cells. Our results revealed that after interaction with the ECM produced by a high metastatic breast cancer cell, MCF-7 cells lost their characteristic epithelial phenotype undergoing EMT, an effect modulated by integrin signaling in crosstalk with TGF-ß receptor signaling pathway. The data evidenced novel potential targets for antimetastatic breast cancer therapies.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Integrina alfaVbeta3/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Integrina alfaVbeta3/genética , Unión Proteica , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
14.
Mol Cell Endocrinol ; 507: 110772, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32114022

RESUMEN

Previous research suggests that omega-3 fatty acids from animal origin may promote the browning of subcutaneous white adipose tissue. We evaluated if supplementation with a plant oil (chia, Salvia hispanica L.) rich in alpha-linolenic fatty acid (C18:3; ω-3) would promote browning and improve glucose metabolism in animals subjected to an obesogenic diet. Swiss male mice (n = 28) were divided into 4 groups: C: control diet; H: high-fat diet; HC: animals in the H group supplemented with chia oil after reaching obesity; HCW: animals fed since weaning on a high-fat diet supplemented with chia oil. Glucose tolerance, inflammatory markers, and expression of genes and proteins involved in the browning process were examined. When supplemented since weaning, chia oil improved glucose metabolism and promoted the browning process and a healthier phenotype. Results of this study suggested that chia oil has potential to protect against the development of obesity-related diseases.


Asunto(s)
Tejido Adiposo Blanco/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Dieta Alta en Grasa , Obesidad/fisiopatología , Aceites de Plantas/farmacología , Salvia/química , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/fisiología , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Tejido Adiposo Blanco/fisiología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Suplementos Dietéticos , Leptina/sangre , Ácidos Linolénicos/farmacología , Masculino , Ratones , Ratones Obesos , Obesidad/dietoterapia , Obesidad/etiología , Obesidad/metabolismo , Aceites de Plantas/aislamiento & purificación
15.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118604, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31760090

RESUMEN

Macrophages (MO) are versatile cells, assuming distinct functional phenotypes depending on the activating stimulus and the microenvironment. The differential activation of macrophages is supported by profound intracellular metabolic changes, being well accepted that the M1/M(LPS+IFN-γ) phenotype rely on aerobic glycolysis, while M2/M(IL-4) macrophages depend on oxidative metabolism. On the other hand, although tumor-associated macrophages (TAMs) are characterized by their high expression of M2/M(IL-4) markers, is currently unclear whether TAMs present the same oxidative metabolic profile of M2/M(IL-4) cells. Herein, we demonstrate for the first time that despite their high expression of M2/M(IL-4) markers, TAMs show high glycolytic activity, with high lactate secretion similar to the M1/M(LPS+ IFN-γ) phenotype. This activity seems to be essential for the M2 profile of TAMs, since the inhibition of glycolysis, but not the impairment of the oxidative phosphorylation or pentose phosphate pathway, diminished the expression of M2/M(IL-4) markers. These novel data indicate that TAMs, although are usually phenotyped as M2/M(IL-4)-like macrophages, they are metabolically distinct from these cells, being rather similar to M1/M(LPS+IFN-γ) macrophages, depending on the glycolytic metabolism to support their profile and functions.


Asunto(s)
Glucólisis , Macrófagos/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Interleucina-4/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Melanoma/metabolismo , Melanoma/patología , Monocitos/citología , Fosforilación Oxidativa/efectos de los fármacos , Vía de Pentosa Fosfato/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
16.
Front Oncol ; 9: 540, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31275860

RESUMEN

During tumor development, the spleen acts as an extra-medullar reservoir of LY6Chi inflammatory monocytes, which can migrate toward tumor to differentiate into tumor-associated macrophage (TAMs), renewing the TAM population. In the tumor microenvironment, pro-inflammatory macrophages (M1) acquire anti-inflammatory and pro-tumor (M2) characteristics favoring tumor development. We previously demonstrated that lipoxins, a family of pro-resolving lipid mediators, restored in vitro the cytotoxic M1-like properties of TAMs. Objective: In this study, we have investigated in vivo the cellular mechanisms underlying the anti-tumor property of lipoxins. Methods: Fourteen days after inducing B16-F10 melanoma tumors, mice received one single dose of ATL-1 (1 µg/i.v.), a lipoxin A4 analog. After further 7 days, blood and bone-marrow were collected, tumors and spleens were removed, and TAMs and blood monocytes were isolated. Results: While the population of LY6Chi monocytes was increased in non-treated tumor-bearing mice, the treatment with ATL-1 diminished the population of LY6Chi monocytes in spleen, blood and bone marrow, decreasing macrophage infiltration into the tumor and reducing the M2 markers expression on TAMs. Importantly, those effects were accompanied by an impairment of tumor growth and improved survival of tumor-bearing mice. The data evidence the anti-tumor mechanism of ATL-1, by decreasing the availability of TAM-precursor monocytes and changing TAMs profile in vivo, impairing tumor progression. ATL-1 may become a new tool in cancer control.

17.
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
18.
Nutrition ; 58: 167-174, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30502680

RESUMEN

OBJECTIVE: Chia seed oil is the richest source of plant-based ω-3 fatty acid, α-linolenic acid, but its potential and mechanisms of action to treat obesity are unclear. The aim of the study was to evaluate the effects of chia oil (ChOi) supplementation on body composition and insulin signaling in skeletal muscles of obese mice. METHODS: Male C57 BL/6 mice (n = 8/group) were fed regular control chow or a high-fat diet (HFD) for 135 d. Another HFD group additionally received ChOi from 90 to 135 d. RESULTS: Consumption of ChOi reduced fat mass accumulation and increased lean mass as evidenced by nuclear magnetic resonance. Moreover, obese mice treated with ChOi showed higher tyrosine phosphorylation of insulin receptor substrate 1, greater activation of protein kinase B, and increased translocation of glucose transporter type 4 in skeletal muscle tissue in response to insulin. ChOi supplementation improved glucose levels and insulin tolerance; decreased serum insulin, leptin, and triacylglycerols; and increased blood high-density lipoprotein cholesterol levels. All these effects caused by the use of ChOi seemed to be independent of the resolution of inflammation because the markers of inflammation were not altered in animals fed the HFD. CONCLUSION: The molecular effects observed in muscle tissue together with changes in body composition may have contributed to the increased glucose tolerance and to the healthy phenotype presented by obese animals treated with ChOi.


Asunto(s)
Composición Corporal/efectos de los fármacos , Suplementos Dietéticos , Insulina/metabolismo , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Aceites de Plantas/farmacología , Salvia , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Músculo Esquelético/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
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.
Int J Mol Sci ; 21(1)2019 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-31905708

RESUMEN

: Nanodrugs have in recent years been a subject of great debate. In 2017 alone, almost 50 nanodrugs were approved for clinical use worldwide. Despite the advantages related to nanodrugs/nanomedicine, there is still a lack of information regarding the biological safety, as the real behavior of these nanodrugs in the body. In order to better understand these aspects, in this study, we evaluated the effect of polylactic acid (PLA) nanoparticles (NPs) and magnetic core mesoporous silica nanoparticles (MMSN), of 1000 nm and 50 nm, respectively, on human cells. In this direction we evaluated the cell cycle, cytochemistry, proliferation and tubulogenesis on tumor cells lines: from melanoma (MV3), breast cancer (MCF-7, MDA-MB-213), glioma (U373MG), prostate (PC3), gastric (AGS) and colon adenocarcinoma (HT-29) and non-tumor cell lines: from human melanocyte (NGM), fibroblast (FGH) and endothelial (HUVEC), respectively. The data showed that an acute exposure to both, polymeric nanoparticles or MMSN, did not show any relevant toxic effects on neither tumor cells nor non-tumor cells, suggesting that although nanodrugs may present unrevealed aspects, under acute exposition to human cells they are harmless.


Asunto(s)
Nanopartículas/toxicidad , Ciclo Celular , Proliferación Celular , Óxido Ferrosoférrico/química , Fibroblastos/metabolismo , Fibroblastos/fisiología , Células HT29 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Células MCF-7 , Nanopartículas/química , Poliésteres/química , Dióxido de Silicio/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA