Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Antioxidants (Basel) ; 13(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39061870

ABSTRACT

While cytostatic chemotherapy targeting DNA is known to induce genotoxicity, leading to cell cycle arrest and cytokine secretion, the impact of these drugs on fibroblast-epithelial cancer cell communication and metabolism remains understudied. Our research focused on human breast fibroblast RMF-621 exposed to nonlethal concentrations of cisplatin and doxorubicin, revealing reduced proliferation, diminished basal and maximal mitochondrial respirations, heightened mitochondrial ROS and lactate production, and elevated MCT4 protein levels. Interestingly, RMF-621 cells enhanced glucose uptake, promoting lactate export. Breast cancer cells MCF-7 exposed to conditioned media (CM) from drug-treated stromal RMF-621 cells increased MCT1 protein levels, lactate-driven mitochondrial respiration, and a significantly high mitochondrial spare capacity for lactate. These changes occurred alongside altered mitochondrial respiration, mitochondrial membrane potential, and superoxide levels. Furthermore, CM with doxorubicin and cisplatin increased migratory capacity in MCF-7 cells, which was inhibited by MCT1 (BAY-8002), glutamate dehydrogenase (EGCG), mitochondrial pyruvate carrier (UK5099), and complex I (rotenone) inhibitors. A similar behavior was observed in T47-D and ZR-75-1 breast cancer cells. This suggests that CM induces metabolic rewiring involving elevated lactate uptake to sustain mitochondrial bioenergetics during migration. Treatment with the mitochondrial-targeting antioxidant mitoTEMPO in RMF-621 and the addition of an anti-CCL2 antibody in the CM prevented the promigratory MCF-7 phenotype. Similar effects were observed in THP1 monocyte cells, where CM increased monocyte recruitment. We propose that nonlethal concentrations of DNA-damaging drugs induce changes in the cellular environment favoring a promalignant state dependent on mitochondrial bioenergetics.

2.
J Periodontal Res ; 59(3): 611-621, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38500269

ABSTRACT

BACKGROUND AND OBJECTIVE: Forkhead box-O 1 (FOXO1) is a transcription factor actively involved in oral wound healing at the epithelial barrier. However, less is known regarding the role of FOXO1 during the tissue repair response in the connective tissue compartment. This study explored the involvement of FOXO1 in the modulation of fibroblast activity related to wound healing. METHODS: Primary cultures of human gingival fibroblasts were obtained from four healthy young donors. Myofibroblastic differentiation, collagen gel contraction, cell migration, cell spreading, and integrin activation were evaluated in the presence or absence of a FOXO1 inhibitor (AS1842856). Variations in mRNA and proteins of interest were evaluated through qRT-PCR and western blot, respectively. Distribution of actin, α-smooth muscle actin, and ß1 integrin was evaluated using immunofluorescence. FOXO1 and TGF-ß1 expression in gingival wound healing was assessed by immunohistochemistry in gingival wounds performed in C57BL/6 mice. Images were analyzed using ImageJ/Fiji. ANOVA or Kruskal-Wallis test followed by Tukey's or Dunn's post-hoc test was performed. All data are expressed as mean ± SD. p < .05 was considered statistically significant. RESULTS: FOXO1 inhibition caused a decrease in the expression of the myofibroblastic marker α-SMA along with a reduction in fibronectin, type I collagen, TGF-ß1, and ß1 integrin mRNA level. The FOXO1 inhibitor also caused decreases in cell migration, cell spreading, collagen gel contraction, and ß1 integrin activation. FOXO1 and TGF-ß1 were prominently expressed in gingival wounds in fibroblastic cells located at the wound bed. CONCLUSION: The present study indicates that FOXO1 plays an important role in the modulation of several wound-healing functions in gingival fibroblast. Moreover, our findings reveal an important regulatory role for FOXO1 on the differentiation of gingival myofibroblasts, the regulation of cell migration, and collagen contraction, all these functions being critical during tissue repair and fibrosis.


Subject(s)
Actins , Cell Movement , Fibroblasts , Forkhead Box Protein O1 , Gingiva , Wound Healing , Humans , Gingiva/cytology , Gingiva/metabolism , Wound Healing/physiology , Fibroblasts/metabolism , Forkhead Box Protein O1/metabolism , Animals , Cells, Cultured , Cell Differentiation , Mice, Inbred C57BL , Transforming Growth Factor beta1/metabolism , Mice , Integrin beta1 , Myofibroblasts , Quinolones
3.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166749, 2023 08.
Article in English | MEDLINE | ID: mdl-37196859

ABSTRACT

BACKGROUND: Metabolic alterations associated with obesity have been related to chronodisruption i.e., the desynchronization of molecular clocks that regulate circadian rhythms. The search for tools that improve the dietary treatment of obesity has recently focused on behaviors related to chronodisruption, and intermittent fasting is increasingly gaining interest. Studies in animal models have identified the benefits of time-restricted feeding (TRF) on metabolic alterations associated with changes in circadian rhythms induced by a high-fat diet. We aimed to evaluate the effect of TRF in flies with metabolic damage and chronodisruption. METHODS: Using high-fat diet fed Drosophila melanogaster as a model of metabolic damage and chronodisruption, we determined the impact of 12-h TRF on metabolic and molecular markers. Flies with metabolic dysfunction were switched to a control diet and randomly assigned to Ad libitum or a TRF regimen for seven days. We evaluated total triglyceride content, glycemia, weight, and 24 h mRNA expression rhythms of Nlaz (insulin resistance marker), clock genes (circadian rhythm molecular markers), and the neuropeptide Cch-amide2. RESULTS: Flies with metabolic damage that received TRF showed lower total triglyceride content, Nlaz expression, circulating glucose, and weight compared to Ad libitum. We observed the recovery of some of the high-fat diet-induced alterations in the amplitude of the circadian rhythm, particularly in the peripheral clock. CONCLUSIONS: TRF produced a partial reversal of metabolic dysfunction and chronodisruption of circadian cycles. GENERAL SIGNIFICANCE: TRF could be a useful tool to help to ameliorate metabolic and chronobiologic damage induced by a high-fat diet.


Subject(s)
Diet, High-Fat , Insulin Resistance , Animals , Diet, High-Fat/adverse effects , Drosophila melanogaster , Obesity/metabolism , Circadian Rhythm/physiology
4.
Antioxidants (Basel) ; 12(4)2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37107188

ABSTRACT

The influence of an inflammatory microenvironment on tumorigenesis has been widely accepted. Systemic conditions that favor the onset of an inflammatory landscape predispose the progression of breast cancer. Under obesity conditions, the endocrine function of adipose tissue is one of the main determinants of the production of local and systemic inflammatory mediators. Although these mediators can stimulate tumorigenesis and recruit inflammatory cells, as macrophages, the mechanism involved remains poorly understood. In the present work, we describe that the TNFα treatment of mammary preadipocytes from human normal patients blocks adipose differentiation and promotes the generation of pro-inflammatory soluble factors. The latter stimulate the mobilization of THP-1 monocytes and MCF-7 epithelial cancer cells in an MCP1/CCL2- and mitochondrial-ROS-dependent manner. Together, these results reaffirm the contribution of an inflammatory microenvironment and mtROS in the progression of breast cancer.

5.
Metabolites ; 11(7)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34357326

ABSTRACT

Breast tumors belong to the type of desmoplastic lesion in which a stiffer tissue structure is a determinant of breast cancer progression and constitutes a risk factor for breast cancer development. It has been proposed that cancer-associated stromal cells (responsible for this fibrotic phenomenon) are able to metabolize glucose via lactate production, which supports the catabolic metabolism of cancer cells. The aim of this work was to investigate the possible functional link between these two processes. To measure the effect of matrix rigidity on metabolic determinations, we used compliant elastic polyacrylamide gels as a substrate material, to which matrix molecules were covalently linked. We evaluated metabolite transport in stromal cells using two different FRET (Fluorescence Resonance Energy Transfer) nanosensors specific for glucose and lactate. Cell migration/invasion was evaluated using Transwell devices. We show that increased stiffness stimulates lactate production and glucose uptake by mammary fibroblasts. This response was correlated with the expression of stromal glucose transporter Glut1 and monocarboxylate transporters MCT4. Moreover, mammary stromal cells cultured on stiff matrices generated soluble factors that stimulated epithelial breast migration in a stiffness-dependent manner. Using a normal breast stromal cell line, we found that a stiffer extracellular matrix favors the acquisition mechanistical properties that promote metabolic reprograming and also constitute a stimulus for epithelial motility. This new knowledge will help us to better understand the complex relationship between fibrosis, metabolic reprogramming, and cancer malignancy.

6.
Int J Obes (Lond) ; 45(7): 1464-1475, 2021 07.
Article in English | MEDLINE | ID: mdl-33895783

ABSTRACT

BACKGROUND: Excess hepatic triglyceride (TG) accumulation (steatosis) commonly observed in obesity, may lead to non-alcoholic fatty liver disease (NAFLD). Altered regulation of intracellular lipid droplets (LD) and TG metabolism, as well as activation of JNK-mediated proinflammatory pathways may trigger liver steatosis-related disorders. Drosophila melanogaster is an animal model used for studying obesity and its associated disorders. In Drosophila, lipids and glycogen are stored in the fat body (FB), which resembles mammalian adipose tissue and liver. Dietary oversupply leads to obesity-related disorders, which are characterized by FB dysfunction. Infusions of Lampaya medicinalis Phil. (Verbenaceae) are used in folk medicine of Chile to counteract inflammatory diseases. Hydroethanolic extract of lampaya (HEL) contains considerable amounts of flavonoids that may explain its anti-inflammatory effect. METHODS: We studied whether HEL affects palmitic acid (PA, C16:0) and oleic acid (OA; C18:1)-induced TG accumulation and proinflammatory marker content in HepG2 hepatocytes as well as impaired lipid storage and proinflammatory molecule expression in Drosophila melanogaster fed a high-fat diet (HFD). RESULTS: In HepG2 hepatocytes, exposure to OA/PA elevated TG content, FABP4, ATGL and DGAT2 expression, and the JNK proinflammatory pathway, as well as TNF-α and IL-6 production, while diminished FAS expression. These effects were prevented by HEL co-treatment. In Drosophila larvae fed a HFD, HEL prevented TG accumulation and downregulated proinflammatory JNK pathway activation. CONCLUSION: HEL effect counteracting OA/PA- and HFD-induced lipid accumulation and proinflammatory marker expression in HepG2 hepatocytes and Drosophila larvae may represent a preventive approach against hepatic steatosis and inflammation, associated to obesity and NAFLD.


Subject(s)
Adipose Tissue/drug effects , Diet, High-Fat/adverse effects , Plant Extracts/pharmacology , Triglycerides/metabolism , Verbenaceae/chemistry , Animals , Drosophila melanogaster , Fat Body/drug effects , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inflammation/metabolism
7.
Sci Rep ; 11(1): 3263, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547367

ABSTRACT

Insulin resistance is a hallmark of type 2 diabetes resulting from the confluence of several factors, including genetic susceptibility, inflammation, and diet. Under this pathophysiological condition, the dysfunction of the adipose tissue triggered by the excess caloric supply promotes the loss of sensitivity to insulin at the local and peripheral level, a process in which different signaling pathways are involved that are part of the metabolic response to the diet. Besides, the dysregulation of insulin signaling is strongly associated with inflammatory processes in which the JAK/STAT pathway plays a central role. To better understand the role of JAK/STAT signaling in the development of insulin resistance, we used a simple organism, Drosophila melanogaster, as a type 2 diabetes model generated by the consumption of a high-sugar diet. In this model, we studied the effects of inhibiting the expression of the JAK/STAT pathway receptor Domeless, in fat body, on adipose metabolism and glycemic control. Our results show that the Domeless receptor loss in fat body cells reverses both hyperglycemia and the increase in the expression of the insulin resistance marker Nlaz, observed in larvae fed a high sugar diet. This effect is consistent with a significant reduction in Dilp2 mRNA expression and an increase in body weight compared to wild-type flies fed high sugar diets. Additionally, the loss of Domeless reduced the accumulation of triglycerides in the fat body cells of larvae fed HSD and also significantly increased the lifespan of adult flies. Taken together, our results show that the loss of Domeless in the fat body reverses at least in part the dysmetabolism induced by a high sugar diet in a Drosophila type 2 diabetes model.


Subject(s)
Diet, Carbohydrate Loading/adverse effects , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Fat Body/metabolism , Insulin Resistance , Receptors, Interleukin/genetics , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Deletion , Insulin/metabolism , Receptors, Interleukin/metabolism
8.
J Periodontal Res ; 55(3): 432-440, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31943227

ABSTRACT

BACKGROUND AND OBJECTIVE: Aging is characterized by a decline in tissue structure and function that may be explained by the development of cellular senescence. However, the acquisition of specific phenotypic responses in senescent gingival fibroblasts is still poorly understood. Here, we have analyzed whether proliferation of primary cultures of human gingival fibroblasts may affect different cell functions relevant to cellular senescence and tissue deterioration. METHODS: Human gingival fibroblasts from five young donors were expanded until cellular senescence was achieved. Cellular senescence was evaluated by determining modifications in cell size, cell proliferation, p16 and p21 mRNA levels, H2Ax phosphorylation, cell viability, and senescence-associated beta-galactosidase staining. Inflammation was evaluated by analyzing the secretion of cytokines and nuclear translocation of NF-κB. Collagen remodeling was evaluated using a collagen gel contraction assay. Immunofluorescence and confocal microscopy were used to determine changes in the localization of the cytoskeletal proteins. Data analysis was performed to identify changes between cultures of the same donor at early and late passages using the paired sample t test or the Wilcoxon matched-pairs signed-rank test. RESULTS: Late passage cells showed changes compatible with cellular senescence that included increased cell size, reduced cell proliferation, staining for SA-beta gal, phosphorylated H2Ax, and increased p16 and p21 mRNA levels. Late passage cells showed a decrease in collagen contraction and reduced co-localization between the cytoskeletal proteins actin and vinculin. Importantly, late passage cells neither demonstrated changes in the secretion of inflammatory cytokines nor NF-κB activation. CONCLUSION: Our results imply that cytoskeletal changes and inhibition of cell proliferation represent early modifications in the structure and function of senescent gingival fibroblasts that are not coupled with the acquisition of an inflammatory phenotype. Further studies are needed to clarify the impact of different senescence stages during aging of the periodontium.


Subject(s)
Cell Proliferation , Cellular Senescence , Cytoskeleton/physiology , Fibroblasts/cytology , Aging , Cells, Cultured , Gingiva/cytology , Humans
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3585-3594, 2018 11.
Article in English | MEDLINE | ID: mdl-30251678

ABSTRACT

Obesity is a major current public health problem worldwide due to the severe co-morbid conditions that this disease entails. The development of obesity-related cardiometabolic disorders is in direct association with adipose tissue inflammation that leads to its functional impairment. Activation of the Calcium-Sensing Receptor (CaSR) in adipose tissue contributes to inflammation and adipose dysfunction. Autophagy, a process of cell component degradation, is closely related to inflammation in many diseases, however, whether autophagy is associated with CaSR-induced inflammation remains unknown. Using LS14 and SW872 preadipose cell lines as well as primary human preadipocytes, we show that CaSR activation with the allosteric activator cinacalcet induces autophagosome formation. Cinacalcet-induced LC3II content elevation was precluded by knockdown of the CaSR and enhanced by CaSR overexpression, indicating a specific effect. Autophagy inhibition using 3-methyladenine prevented CaSR-induced TNFα production, indicating that autophagy contributes to CaSR-induced inflammation in human preadipocytes. Our results suggest that modulation of CaSR-induced autophagy is an attractive target in obese inflamed adipose tissue, to prevent the development of diseases triggered by adipose dysfunction. We describe a novel mechanism and possible new target to modulate and prevent adipose inflammation and hence the resulting disease-generating adipose tissue dysfunction.


Subject(s)
Adipose Tissue/pathology , Autophagy , Inflammation/pathology , Receptors, Calcium-Sensing/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Calcimimetic Agents/pharmacology , Cell Line , Cinacalcet/pharmacology , Gene Knockdown Techniques , Humans , Inflammation/etiology , Obesity/complications , Obesity/metabolism , Primary Cell Culture , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/genetics
10.
Redox Biol ; 16: 199-208, 2018 06.
Article in English | MEDLINE | ID: mdl-29524842

ABSTRACT

Aerobic metabolism brings inexorably the production of reactive oxygen species (ROS), which are counterbalanced by intrinsic antioxidant defenses avoiding deleterious intracellular effects. Redox balance is the resultant of metabolic functioning under environmental inputs (i.e. diet, pollution) and the activity of intrinsic antioxidant machinery. Monitoring of intracellular hydrogen peroxide has been successfully achieved by redox biosensor advent; however, to track the intrinsic disulfide bond reduction capacity represents a fundamental piece to understand better how redox homeostasis is maintained in living cells. In the present work, we compared the informative value of steady-state measurements and the kinetics of HyPer, a H2O2-sensitive fluorescent biosensor, targeted at the cytosol, mitochondrion and endoplasmic reticulum. From this set of data, biosensor signal recovery from an oxidized state raised as a suitable parameter to discriminate reducing capacity of a close environment. Biosensor recovery was pH-independent, condition demonstrated by experiments on pH-clamped cells, and sensitive to pharmacological perturbations of enzymatic disulfide reduction. Also, ten human cell lines were characterized according their H2O2-pulse responses, including their capacity to reduce disulfide bonds evaluated in terms of their migratory capacity. Finally, cellular migration experiments were conducted to study whether migratory efficiency was associated with the disulfide reduction activity. The migration efficiency of each cell type correlates with the rate of signal recovery measured from the oxidized biosensor. In addition, HyPer-expressing cells treated with N-acetyl-cysteine had accelerated recovery rates and major migratory capacities, both reversible effects upon treatment removal. Our data demonstrate that the HyPer signal recovery offers a novel methodological tool to track the cellular impact of redox active biomolecules.


Subject(s)
Antioxidants/isolation & purification , Biosensing Techniques , Hydrogen Peroxide/chemistry , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mitochondria/metabolism , Oxidation-Reduction
11.
J Cell Biochem ; 118(5): 994-1002, 2017 05.
Article in English | MEDLINE | ID: mdl-27403856

ABSTRACT

Chronic inflammation and metabolic reprogramming have been proposed as hallmarks of cancer development. Currently, many of the functional clues between these two phenomena are studied under the integrative view of functional stroma-epithelia interaction. It has been proposed that stromal cells, due to their abundance and avidity for glucose, are able to modify the metabolic behavior of an entire solid tumor. In the present study, using a mammary stromal cell line derived from healthy tissue subjected to long-term culture in low (5 mM) or high (25 mM) glucose, we found that the hyperglycemic condition favors the establishment of a pro-inflammatory and pro-oxidant environment characterized by the induction of the COX-2/PGE2 axis. In this condition, epithelial migration was stimulated. Moreover, we also found that stromal-derived PGE2, acting as a stimulator of IL-1 epithelial expression was one of the factors that promote the acquisition of motile properties by epithelial cells and the maintenance of a COX-2/PGE2-dependent inflammatory condition. Overall, our work provides experimental evidence that glucose stimulates a tumor inflammatory environment that, as a result of a functional cross-talk between stroma and epithelia, may be responsible for tumor progression. J. Cell. Biochem. 118: 994-1002, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Breast Neoplasms/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/pharmacology , Glucose/pharmacology , Interleukin-1/metabolism , Stromal Cells/cytology , Breast Neoplasms/immunology , Cell Culture Techniques , Cell Line, Tumor , Cell Movement , Cells, Cultured , Female , Humans , MCF-7 Cells , Reactive Oxygen Species/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Microenvironment
12.
J Cell Physiol ; 232(1): 136-44, 2017 01.
Article in English | MEDLINE | ID: mdl-27037895

ABSTRACT

Hyperglycemia is a risk factor for a variety of human cancers. Increased access to glucose and that tumor metabolize glucose by a glycolytic process even in the presence of oxygen (Warburg effect), provide a framework to analyze a particular set of metabolic adaptation mechanisms that may explain this phenomenon. In the present work, using a mammary stromal cell line derived from healthy tissue that was subjected to a long-term culture in low (5 mM) or high (25 mM) glucose, we analyzed kinetic parameters of lactate transport using a FRET biosensor. Our results indicate that the glucose pre-culture and soluble epithelial factors constitute a stimulus for lactate stromal production, factors that also modify the kinetic parameters and the monocarboxylate transporters expression in stromal cells. We also observed a vectorial flux of lactate from stroma to epithelial cells in a co-culture setting and found that the uptake of lactate by epithelial cells correlates with the degree of malignancy. Glucose preconditioning of the stromal cell stimulated epithelial motility. Our findings suggest that lactate generated by stromal cells in the high glucose condition stimulate epithelial migration. Overall, our results support the notion that glucose not only provides a substrate for tumor nutrition but also behaves as a signal promoting malignancy. J. Cell. Physiol. 232: 136-144, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Movement/physiology , Epithelial Cells/metabolism , Glucose/metabolism , Stromal Cells/metabolism , Breast/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Coculture Techniques , Humans , Lactic Acid/metabolism , Mammary Glands, Human/metabolism , Mitochondria/metabolism
13.
Rev. Hosp. Clin. Univ. Chile ; 27(1): 12-20, 2016. tab, graf
Article in Spanish | LILACS | ID: biblio-908176

ABSTRACT

Introduction: eating disorders(ED)are chronic mental illnesses, highly prevalent within adolescent population, affecting women more than men. Generally, ED are triggered by an unsatisfactory self body-image, which relates to several socio-cultural factors. Objective: To study the prevalence of the risk of developing ED within Chilean high school students, and to evaluate its relation with sex and the school gender composition (SGC): single-sex or coeducational. Method: A transversal-analytic study was performed using three Santiago public high schools: female-exclusive, male-exclusive and coeducational. The Eating Attitudes Test-40 was applied, considering a 30-point score to define qualitatively the risk condition. Results: The sample included 415 students, 52 percent women, with a mean age of 15,9 years (range 14 to 18). The global prevalence of risk for ED was 14,9 percent. The risk condition was significantly higher in women (25,2 percent) than men (4 percent). No differences were found when comparing the prevalence of risk according to the type of school. Discussion: The higher risk for ED among women respect to men was confirmed, and the SGC has no influence on this risk.


Subject(s)
Male , Female , Humans , Adolescent , Feeding and Eating Disorders/epidemiology , Feeding and Eating Disorders/psychology , Sex Factors , Students , Chile , Cross-Sectional Studies , Feeding Behavior , Risk Assessment , Sex Distribution
14.
BMC Cancer ; 14: 640, 2014 Aug 30.
Article in English | MEDLINE | ID: mdl-25175743

ABSTRACT

BACKGROUND: Hard consistency, developed under the influence of tumor cell factors, is a characteristic feature of a breast tumor. Activation of resident fibroblasts leading to a myofibroblast phenotype is the principal feature that orchestrates this fibrotic process. The aim of this study was to assess the effects induced by TGF-ß1, a growth factor abundantly present in tumor microenvironment, on the molecular mechanisms that mediate myofibroblastic differentiation of normal human mammary fibroblasts. METHODS: We used an immortalized fibroblastic cell line derived from normal mammary tissue (RMF-EG cells) to study the effect of TGF-ß1 in the expression of α-SMA and CTGF as markers of myofibroblastic differentiation. The influence of redox status and JNK activity on TGF-ß1-induced transcriptional activity was measured by a luciferase reporter assay. We also used a shRNA approach to evaluate the influence of NOX4 in myofibroblastic differentiation. RESULTS: TGF-ß1 stimulates the expression of myofibroblast markers α-SMA and CTGF. Using a NOX inhibitor (DPI) and cells expressing a shRNA for NOX4, we demonstrated that TGF-ß1 promotes an oxidative environment that favors myofibroblastic differentiation. We also found that activation of c-Jun N-terminal kinase is required for TGF-ß1-dependent expression of CTGF, NOX4 and α-SMA. CONCLUSIONS: Human mammary stromal fibrosis, evaluated by the expression of early and late markers as CTGF and α-SMA, depends on the activation of JNK signaling pathway. Our results show that JNK activation is an early event that precedes the increase in ROS levels leading to myofibroblastic differentiation and tumor fibrosis, suggesting that inhibition of JNK may be used a method to interrupt the development of tumor desmoplasia.


Subject(s)
Breast/cytology , JNK Mitogen-Activated Protein Kinases/metabolism , Myofibroblasts/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/metabolism , Actins/metabolism , Breast/metabolism , Breast/pathology , Cell Differentiation , Cell Line , Connective Tissue Growth Factor/metabolism , Female , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , MAP Kinase Signaling System/drug effects , NADPH Oxidase 4 , Stromal Cells/metabolism
15.
Carcinogenesis ; 35(8): 1770-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24618373

ABSTRACT

It has been proposed that epithelial cells can acquire invasive properties through exposure to paracrine signals originated from mesenchymal cells within the tumor microenvironment. Transforming growth factor-ß (TGF-ß) has been revealed as an active factor that mediates the epithelial-stroma cross-talk that facilitates cell invasion and metastasis. TGF-ß signaling is modulated by the coreceptor Endoglin (Eng), which shows a tumor suppressor activity in epithelial cells and regulates the ALK1-Smad1,5,8 as well as the ALK5-Smad2,3 signaling pathways. In the current work, we present evidence showing that cell surface Eng abundance in epithelial MCF-7 breast cancer cells is inversely related with cell motility. Shedding of Eng in MCF-7 cell surface by soluble matrix metalloproteinase-14 (MMP-14) derived from the HS-5 bone-marrow-derived cell line induces a motile epithelial phenotype. On the other hand, restoration of full-length Eng expression blocks the stromal stimulus on migration. Processing of surface Eng by stromal factors was demonstrated by biotin-neutravidin labeling of cell surface proteins and this processing generated a shift in TGF-ß signaling through the activation of Smad2,3 pathway. Stromal MMP-14 abundance was stimulated by TGF-ß secreted by MCF-7 cells acting in a paracrine manner. In turn, the stromal proteolytic activity of soluble MMP-14, by inducing Eng shedding, promoted malignant progression. From these data, and due to the capacity of TGF-ß to regulate malignancy in epithelial cancer, we propose that stromal-dependent epithelial Eng shedding constitutes a putative mechanism that exerts an environmental control of cell malignancy.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Epithelial Cells/pathology , Matrix Metalloproteinase 14/metabolism , Mesenchymal Stem Cells/pathology , Receptors, Cell Surface/metabolism , Antigens, CD/genetics , Blotting, Western , Breast Neoplasms/genetics , Cell Proliferation , Culture Media, Conditioned/pharmacology , Endoglin , Epithelial Cells/metabolism , Female , Flow Cytometry , Humans , Immunoprecipitation , Matrix Metalloproteinase 14/genetics , Mesenchymal Stem Cells/metabolism , Microscopy, Fluorescence , Phosphorylation/drug effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
16.
Mol Cell Biochem ; 384(1-2): 139-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24005534

ABSTRACT

We established that human adipose cells and the human adipose cell line LS14 express the calcium-sensing receptor (CaSR) and that its activation induces inflammatory cytokine production. Also, its expression is enhanced upon exposure to obesity-associated proinflammatory cytokines. We have thus proposed that CaSR activation may be associated with adipose dysfunction. Here, we evaluated a possible effect on adipogenesis. We induced adipose differentiation of primary and LS14 human preadipocytes with or without the simultaneous activation of CaSR, by the exposure to the calcimimetic cinacalcet. Activation of the receptor for 24 h decreased by 40 % the early differentiation marker CCAAT/enhancer-binding protein ß. However, upon longer-term (10 day) exposure to the adipogenic cocktail, cinacalcet exerted the opposite effect, causing a dose-response increase in the expression of the mature adipose markers adipocyte protein 2, adiponectin, peroxisome proliferator-activated receptor γ, fatty acid synthase, and glycerol-3-phosphate dehydrogenase. To assess whether there was a time-sensitive effect of CaSR activation on adipogenesis, we evaluated the 10 day effect of cinacalcet exposure for the first 6, 24, 48 h, 6, and 10 days. Our observations suggest that regardless of the period of exposure, 10 day adipogenesis is elevated by cinacalcet. CaSR activation may interfere with the initial stages of adipocyte differentiation; however, these events do not seem to preclude adipogenesis from continuing. Even though adipogenesis (particularly in subcutaneous depots) is associated with insulin sensitivity and adequate adipose function, the implications of our findings in visceral adipocytes, especially in the context of inflamed AT and overnutrition, remain to be established.


Subject(s)
Adipocytes/metabolism , Adipogenesis/drug effects , Naphthalenes/pharmacology , Receptors, Calcium-Sensing/metabolism , Adipogenesis/physiology , Adiponectin/biosynthesis , Adiponectin/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Cinacalcet , Cytokines/metabolism , Enzyme Activation , Fatty Acid Synthases/biosynthesis , Fatty Acid Synthases/metabolism , Female , Glycerolphosphate Dehydrogenase/biosynthesis , Glycerolphosphate Dehydrogenase/metabolism , Humans , Inflammation , Male , PPAR gamma/biosynthesis , PPAR gamma/metabolism
17.
J Periodontol ; 84(5): 683-93, 2013 May.
Article in English | MEDLINE | ID: mdl-22813343

ABSTRACT

BACKGROUND: Fibroblasts play a critical role during wound healing and chronic inflammation through the synthesis and assembly of extracellular matrix (ECM) molecules. These responses may be modulated by soluble cytokines and growth factors present in tissues. In the present study, we evaluate whether transforming growth factor-ß1 (TGF-ß1) and tumor necrosis factor-α (TNF-α) modulate myofibroblastic differentiation and the production of ECM components. METHODS: Primary cultures of human gingival fibroblasts (HGFs) were stimulated with recombinant TGF-ß1 and TNF-α. Protein levels of α-smooth muscle actin (α-SMA), type I collagen, heat shock protein-47 (HSP-47), fibronectin (FN), ED-A-FN, and periostin and activation of the Smad pathway were evaluated through Western blot analysis. α-SMA and actin fibers were identified by immunofluorescence. TGF-ß1, TNF-α, and α-SMA were identified by immunohistochemistry in biopsies of inflamed human gingival tissues. TGF-ß1 activity was evaluated using a plasminogen activator inhibitor-1 (PAI-1) reporter transfected in HGFs. RESULTS: TGF-ß1 stimulated the differentiation of myofibroblasts as evidenced by strong expression of α-SMA and ED-A-FN. Moreover, TGF-ß1 induced the production of type I collagen, HSP-47, FN, and periostin. Costimulation with TNF-α and TGF-ß1 significantly reduced the expression of all the above-mentioned proteins. TNF-α also inhibited the activation of the Smad2/3 pathway and the activity of the PAI-1 reporter. CONCLUSIONS: TNF-α inhibits several cell responses induced by TGF-ß1, including the differentiation of myofibroblasts, the activation of the Smad signaling pathway, and the production of key molecules involved in tissue repair, such as type I collagen, FN, and periostin. The interaction between cytokines may explain the delayed tissue repair observed in chronic inflammation of gingival tissues.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix/drug effects , Gingiva/metabolism , Myofibroblasts/drug effects , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Actins/biosynthesis , Adult , Cell Adhesion Molecules/biosynthesis , Cell Differentiation/drug effects , Cells, Cultured , Chronic Periodontitis/metabolism , Collagen Type I/biosynthesis , Female , Fibronectins/biosynthesis , Gingiva/cytology , Humans , Male , Middle Aged , Plasminogen Activator Inhibitor 1/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism
18.
Mol Cell Endocrinol ; 361(1-2): 24-30, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22449852

ABSTRACT

The proinflammatory status of adipose tissue has been linked to the metabolic and cardiovascular consequences of obesity. Human adipose cells express the calcium sensing receptor (CaSR), and its expression is elevated in inflammatory states, such as that associated with obesity. Given the CaSR's association with inflammation in other tissues, we evaluated its role elevating the adipose expression of inflammatory factors. The CaSR activation by the calcimimatic cinacalcet (5µM) in adipose tissue and in vitro cultured LS14 adipose cells elicited an elevation in the expression of the proinflammatory cytokines IL6, IL1ß, TNFα, and the chemoattractant CCL2. This was in part reverted by SN50, an inhibitor of the inflammatory mediator nuclear factor kappa B (NFκB). Our observations suggest that CaSR activation elevates cytokine and chemokine production, partially mediated by NFκB. These findings support the relevance of the CaSR in the pathophysiology of obesity-induced adipose tissue dysfunction, with an interesting potential for pharmacological manipulation.


Subject(s)
Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Inflammation Mediators/metabolism , Receptors, Calcium-Sensing/metabolism , Adipocytes/drug effects , Adipose Tissue/drug effects , Adult , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cinacalcet , Cytokines/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Male , Middle Aged , NF-kappa B/metabolism , Naphthalenes/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcium-Sensing/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Young Adult
19.
Arch Biochem Biophys ; 500(2): 151-6, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20595056

ABSTRACT

Obesity-associated health complications are thought to be in part due to the low-grade proinflammatory state that characterizes this disease. The calcium sensing receptor (CaSR), which is expressed in human adipose cells, plays an important role in diseases involving inflammation. To assess the relevance of this protein in adipose pathophysiology, we evaluated its expression in adipocytes under obesity-related proinflammatory conditions. As in primary adipose cells, we established that LS14, a recently described human adipose cell line, expresses the CaSR. Differentiated LS14 and primary adipose cells were exposed overnight to cytokines typically involved in obesity-related inflammation (interleukin (IL)1beta, IL6 and tumor necrosis factor (TNF)alpha). The cytokines increased CaSR abundance in differentiated adipocytes. We incubated LS14 cells with medium previously conditioned (CM) by adipose tissue from subjects with a wide range of body mass index (BMI). Cells exposed to CM from subjects of higher BMI underwent a greater increase in CaSR protein, likely resulting from the greater proinflammatory cytokines secreted from obese tissue. Our observations that proinflammatory factors increase CaSR levels in adipocytes, and the reported ability of CaSR to elevate cytokine levels, open new aspects in the study of obesity inflammatory state pathophysiology, providing a potential novel therapeutic prevention and treatment target.


Subject(s)
Adipocytes/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Obesity/metabolism , Receptors, Calcium-Sensing/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipose Tissue/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , Culture Media, Conditioned , Cytokines/pharmacology , Humans , Inflammation Mediators/pharmacology , Interleukin-1beta/pharmacology , Interleukin-6/pharmacology , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/pharmacology
20.
Mol Cell Biochem ; 340(1-2): 195-202, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20204677

ABSTRACT

TGF-beta1 has been postulated as a pro-oncogenic factor in the late step of the tumoral progression. In transformed cells, TGF-beta1 enhances the capacity to degrade the extracellular matrix, cell invasiveness and epithelial-mesenchymal transition, which are crucial steps for metastasis. Urokinase-type plasminogen activator (uPA) and matrix metalloproteinase-9 (MMP-9) are critical components in cell migration and invasion induced by TGF-beta1, however, the exact mechanism by which TGF-beta1 regulates uPA and MMP-9 is not well elucidated so far. In the present study, we analyzed the role of ROS-NFkappaB, signal as mediator in the cell malignity enhancement by TGF-beta1. We found that TGF-beta1 activates NFkappaB, through Rac1-NOXs-ROS-dependent mechanism. Our results shows that TGF-beta1 stimulation of uPA and MMP-9 expression involve NOXs-dependent ROS and NFkappaB, activation, demonstrated by using DPI, NOXs inhibitor, ROS scavenger N-acetylcysteine and SN50, an NFkb inhibitor. Furthermore, we found that the inhibition of ROS and NFkappaB, abrogates TGF-beta1 stimulation of EMT, cell motility and invasion. Thus, ROS-NFkappaB acts as the crucial signal in TGF-beta1-induced uPA and MMP-9 expression thereby mediating the enhancement of cellular malignity by TGF-beta1.


Subject(s)
Cell Movement , Keratinocytes/enzymology , Matrix Metalloproteinase 9/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor RelA/metabolism , Transforming Growth Factor beta1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Transformed , Cell Movement/drug effects , Cell Transdifferentiation , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Genes, Reporter , Keratinocytes/drug effects , Matrix Metalloproteinase 9/genetics , Mice , Mutation , NADPH Oxidases/metabolism , Phenotype , Promoter Regions, Genetic , Signal Transduction , Time Factors , Transcription Factor RelA/genetics , Transfection , Up-Regulation , Urokinase-Type Plasminogen Activator/genetics , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL