Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892249

ABSTRACT

Mesenchymal stromal cell (MSC)-based advanced therapy medicinal products (ATMPs) are being tried in a vast range of clinical applications. These cells can be isolated from different donor tissues by using several methods, or they can even be derived from induced pluripotent stem cells or embryonic stem cells. However, ATMP heterogeneity may impact product identity and potency, and, consequently, clinical trial outcomes. In this review, we discuss these topics and the need to establish minimal criteria regarding the manufacturing of MSCs so that these innovative therapeutics may be better positioned to contribute to the advancement of regenerative medicine.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Regenerative Medicine , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cell Transplantation/methods , Regenerative Medicine/methods , Animals , Induced Pluripotent Stem Cells/cytology , Cell Differentiation
2.
Front Physiol ; 13: 1077069, 2022.
Article in English | MEDLINE | ID: mdl-36589430

ABSTRACT

Human pluripotent stem cells (PSC) have been used for disease modelling, after differentiation into the desired cell type. Electrophysiologic properties of cardiomyocytes derived from pluripotent stem cells are extensively used to model cardiac arrhythmias, in cardiomyopathies and channelopathies. This requires strict control of the multiple variables that can influence the electrical properties of these cells. In this article, we report the action potential variability of 780 cardiomyocytes derived from pluripotent stem cells obtained from six healthy donors. We analyze the overall distribution of action potential (AP) data, the distribution of action potential data per cell line, per differentiation protocol and batch. This analysis indicates that even using the same cell line and differentiation protocol, the differentiation batch still affects the results. This variability has important implications in modeling arrhythmias and imputing pathogenicity to variants encountered in patients with arrhythmic diseases. We conclude that even when using isogenic cell lines to ascertain pathogenicity to variants associated to arrythmias one should use cardiomyocytes derived from pluripotent stem cells using the same differentiation protocol and batch and pace the cells or use only cells that have very similar spontaneous beat rates. Otherwise, one may find phenotypic variability that is not attributable to pathogenic variants.

3.
Genet Mol Biol ; 44(3): e20200147, 2021.
Article in English | MEDLINE | ID: mdl-34496008

ABSTRACT

Induced pluripotent stem cells (iPSCs) are generated from adult cells that have been reprogrammed to pluripotency. However, in vitro cultivation and genetic reprogramming increase genetic instability, which could result in chromosomal abnormalities. Maintenance of genetic stability after reprogramming is required for possible experimental and clinical applications. The aim of this study was to analyze chromosomal alterations by using the G-banding karyotyping method applied to 97 samples from 38 iPSC cell lines generated from peripheral blood or Wharton's jelly. Samples from patients with long QT syndrome, Jervell and Lange-Nielsen syndrome and amyotrophic lateral sclerosis and from normal individuals revealed the following chromosomal alterations: acentric fragments, chromosomal fusions, premature centromere divisions, double minutes, radial figures, ring chromosomes, polyploidies, inversions and trisomies. An analysis of two samples generated from Wharton's jelly before and after reprogramming showed that abnormal clones can emerge or be selected and generate an altered lineage. IPSC lines may show clonal and nonclonal chromosomal aberrations in several passages (from P6 to P34), but these aberrations are more common in later passages. Many important chromosomal aberrations were detected, showing that G-banding is very useful for evaluating genetic instability with important repercussions for the application of iPSC lines.

4.
Cells ; 9(7)2020 07 07.
Article in English | MEDLINE | ID: mdl-32645832

ABSTRACT

Chagas disease discovered more than a century ago remains an incurable disease. The objective of this work was to investigate the therapeutic potential of cardiomyocytes derived from mouse embryonic stem cells (CM-mESC) in a model of chronic Chagasic cardiomyopathy (CCC). Mouse embryonic stem cells (mESC) were characterized, transduced with luciferase, and submitted to cardiac differentiation. CM-mESC were labeled with superparamagnetic iron oxide particles. To induce CCC, mice were infected with Brazil strain trypomastigotes. At 150 days post-infection (dpi), infected animals were treated with CM-mESC or PBS. Cells were detected by magnetic resonance imaging (MRI) and bioluminescence. Cardiac function was evaluated by MRI and electrocardiogram at 150 and 196 dpi. CCC mice showed significant differences in MRI and ECG parameters compared to non-infected mice. However, no differences were observed in contractile and electrical parameters between cell and PBS injected groups, 45 days after cell transplantation. Cells were detected 24 h after transplantation by MRI. CM-mESC bioluminescence tracking demonstrated over 90% decrease in signal 8 days after treatment. Nevertheless, the Infected + CM-mESC group showed a significant reduction in the percentage of collagen fibers when compared to the Infected + PBS group. In conclusion, CM-mESC therapy was not effective in reversing cardiac functional changes induced by Chagas disease despite some improvement in myocardial fibrosis.


Subject(s)
Cardiomyopathies/metabolism , Cardiomyopathies/therapy , Cell- and Tissue-Based Therapy/methods , Myocytes, Cardiac/physiology , Animals , Cardiomyopathies/diagnostic imaging , Chagas Disease/diagnostic imaging , Chagas Disease/metabolism , Chagas Disease/therapy , Disease Models, Animal , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Flow Cytometry , Humans , Magnetic Resonance Imaging , Male , Mice , Myocytes, Cardiac/metabolism
5.
EXCLI J ; 18: 91-105, 2019.
Article in English | MEDLINE | ID: mdl-30956642

ABSTRACT

The therapeutic potential of Baccharis anomala DC. extracts was evaluated through its cytotoxic and antiproliferative effect and their phenotypic reversion property in activated hepatic stellate cells (HSCs). Baccharis anomala is distributed in Brazil (southeastern and south regions) and used for diuretic effect in folk medicine. Four fractions were obtained from the fractionation of the methanolic extract. Fractions III and IV decreased cell proliferation without increasing cell necrosis markers levels and induced cell cycle arrest in G1 phase. Fraction III induced phenotypic reversion through PPAR-γ activation pathway, while fraction IV did not alter PPAR-α/γ expression levels, suggesting that there is an independent PPAR-α/γ pathway involved. Hydroxybenzoic, chlorogenic and coumaric acids were identified. Fractions III and IV showed antiproliferative effect and ability to induce reversion of activated phenotype of HSCs.

6.
Stem Cell Res Ther ; 9(1): 30, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29402309

ABSTRACT

BACKGROUND: Doxorubicin (Dox) is a chemotherapy drug with limited application due to cardiotoxicity that may progress to heart failure. This study aims to evaluate the role of cardiomyocytes derived from mouse embryonic stem cells (CM-mESCs) in the treatment of Dox-induced cardiomyopathy (DIC) in mice. METHODS: The mouse embryonic stem cell (mESC) line E14TG2A was characterized by karyotype analysis, gene expression using RT-PCR and immunofluorescence. Cells were transduced with luciferase 2 and submitted to cardiac differentiation. Total conditioned medium (TCM) from the CM-mESCs was collected for proteomic analysis. To establish DIC in CD1 mice, Dox (7.5 mg/kg) was administered once a week for 3 weeks, resulting in a cumulative Dox dose of 22.5 mg/kg. At the fourth week, a group of animals was injected intramyocardially with CM-mESCs (8 × 105 cells). Cells were tracked by a bioluminescence assay, and the body weight, echocardiogram, electrocardiogram and number of apoptotic cardiomyocytes were evaluated. RESULTS: mESCs exhibited a normal karyotype and expressed pluripotent markers. Proteomic analysis of TCM showed proteins related to the negative regulation of cell death. CM-mESCs presented ventricular action potential characteristics. Mice that received Dox developed heart failure and showed significant differences in body weight, ejection fraction (EF), end-systolic volume (ESV), stroke volume (SV), heart rate and QT and corrected QT (QTc) intervals when compared to the control group. After cell or placebo injection, the Dox + CM-mESC group showed significant increases in EF and SV when compared to the Dox + placebo group. Reduction in ESV and QT and QTc intervals in Dox + CM-mESC-treated mice was observed at 5 or 30 days after cell treatment. Cells were detected up to 11 days after injection. The Dox + CM-mESC group showed a significant reduction in the percentage of apoptotic cardiomyocytes in the hearts of mice when compared to the Dox + placebo group. CONCLUSIONS: CM-mESC transplantation improves cardiac function in mice with DIC.


Subject(s)
Cardiomyopathies/therapy , Doxorubicin/adverse effects , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/transplantation , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Line , Doxorubicin/therapeutic use , Human Embryonic Stem Cells/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Myocytes, Cardiac/pathology
7.
Stem Cell Res Ther ; 8(1): 289, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29273091

ABSTRACT

BACKGROUND: Sepsis is a severe medical condition that ranks among the top 10 causes of death worldwide and which has permanently high incidence rates. Mesenchymal stem cells (MSCs) have been found to be potent modulators of immune responses. More importantly, there is evidence that MSCs have a beneficial effect on preclinical models of polymicrobial sepsis. However, the changes caused by the MSCs in the effector cells of the host immune system remain unclear. METHODS: A mouse model of sepsis (male C57BL/6 mice) with three experimental groups was used for experiments in vivo: a control group, an untreated septic group, and a septic group treated with MSCs. In vitro experiments were performed using a cell line of pulmonary macrophages (RAW 264.7) co-cultured with MSCs and stimulated with lipopolysaccharide (LPS). RESULTS: In vivo we demonstrated that treatment with MSCs was able to reduce the expression of cyclooxygenase-2 (COX-2) and nuclear factor kappa B (NF-κB), and thereby decrease the production of inflammatory cytokines. In vitro experiments using a co-culture of macrophages with MSCs showed a decrease in COX-2 and NF-κB, and showed that this reduction was directly related to the ability of MSCs to inhibit phosphorylation of ERK, RSK, and p38, enzymes that belong to the family of mitogen-activated protein kinases (MAPKs). CONCLUSIONS: This study demonstrated that MSCs are able to inhibit the MAPK pathway activation, modulating the inflammatory response during sepsis. This understanding that MSCs can remodel the response of host cells and improve the course of sepsis is essential for developing new treatments for this pathology.


Subject(s)
Mesenchymal Stem Cells/metabolism , Sepsis/complications , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Pneumonia/pathology , Sepsis/pathology
9.
Sci Rep ; 7(1): 3255, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28607430

ABSTRACT

Hepatic stellate cells (HSC) play a key role in the development of chronic liver disease (CLD). Liraglutide, well-established in type 2 diabetes, showed anti-inflammatory and anti-oxidant properties. We evaluated the effects of liraglutide on HSC phenotype and hepatic microvascular function using diverse pre-clinical models of CLD. Human and rat HSC were in vitro treated with liraglutide, or vehicle, and their phenotype, viability and proliferation were evaluated. In addition, liraglutide or vehicle was administered to rats with CLD. Liver microvascular function, fibrosis, HSC phenotype and sinusoidal endothelial phenotype were determined. Additionally, the effects of liraglutide on HSC phenotype were analysed in human precision-cut liver slices. Liraglutide markedly improved HSC phenotype and diminished cell proliferation. Cirrhotic rats receiving liraglutide exhibited significantly improved liver microvascular function, as evidenced by lower portal pressure, improved intrahepatic vascular resistance, and marked ameliorations in fibrosis, HSC phenotype and endothelial function. The anti-fibrotic effects of liraglutide were confirmed in human liver tissue and, although requiring further investigation, its underlying molecular mechanisms suggested a GLP1-R-independent and NF-κB-Sox9-dependent one. This study demonstrates for the first time that liraglutide improves the liver sinusoidal milieu in pre-clinical models of cirrhosis, encouraging its clinical evaluation in the treatment of chronic liver disease.


Subject(s)
Hepatic Stellate Cells/drug effects , Incretins/pharmacology , Liraglutide/pharmacology , Liver Cirrhosis/drug therapy , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Humans , Liver/blood supply , Liver Cirrhosis/physiopathology , Male , Microvessels/drug effects , Rats, Wistar
10.
Biometals ; 30(4): 549-558, 2017 08.
Article in English | MEDLINE | ID: mdl-28639108

ABSTRACT

Hepatic fibrosis is an extracellular matrix deposition by hepatic stellate cells (HSC). Fibrosis can be caused by iron, which will lead to hydroxyl radical production and cell damage. Fructose-1,6-bisphosphate (FBP) has been shown to deliver therapeutic effects in many pathological situations. In this work, we aimed to test the effects of FBP in HSC cell line, GRX, exposed to an excess of iron (Fe). The Fe-treatment increased cell proliferation and FBP reversed this effect, which was not due to increased necrosis, apoptosis or changes in cell cycle. Oil Red-O staining showed that FBP successfully increased lipid content and lead GRX cells to present characteristics of quiescent HSC. Fe-treatment decreased PPAR-γ expression and increased Col-1 expression. Both effects were reversed by FBP which also decreased TGF-ß1 levels in comparison to both control and Fe groups. FBP, also, did not present scavenger activity in the DPPH assay. The treatment with FBP resulted in decreased proliferation rate, Col-1 expression and TGF-ß1 release by HSC cells. Furthermore, activated PPAR-γ and increased lipid droplets induce cells to become quiescent, which is a key event to reversion of hepatic fibrosis. FBP also chelates iron showing potential to improve Cell redox state.


Subject(s)
Ferrous Compounds/antagonists & inhibitors , Fructosediphosphates/pharmacology , Hepatic Stellate Cells/drug effects , Iron Chelating Agents/pharmacology , Animals , Biphenyl Compounds/chemistry , Cell Line , Cell Survival/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Ferrous Compounds/pharmacology , Gene Expression Regulation , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Mice , Oxidation-Reduction , PPAR gamma/genetics , PPAR gamma/metabolism , Picrates/chemistry , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
11.
Biomed Pharmacother ; 89: 358-365, 2017 05.
Article in English | MEDLINE | ID: mdl-28242545

ABSTRACT

Hepatocellular carcinoma (HCC) is the most prevalent primary liver tumor that affects the world population. Liver cancer inevitably causes great harms and its treatment is extremely difficult. Its development is related to the existence of chronic liver injury, such as in cirrhosis. Cancer is a disease related to the process of inflammation so, research with anti-inflammatory agents has been performed for the development of anti-tumor drugs. Fructose-1,6-bisphosphate (FBP), a metabolite of the glycolytic route, has shown anti-inflammatory actions. The purpose of this study is to investigate the effect of FBP on HepG2 cells growth and inflammatory parameters. Results showed that FBP decreased the proliferation of HepG2 cells through trypan blue assay, without causing necrosis, shown by the intracellular release of LDH. By flow cytometry, we observed a significant IL-8 decrease which is closely related to the tumoral progression and chemotherapeutic resistance, especially in HCC. Then, we found, by RT-PCR, a high expression level of pro-apoptotic protein, such as Bax and p53, and decreased the expression levels of anti-apoptotic proteins, like Bcl-2 suggesting apoptosis. Finally, our results showed that FBP can be a potential therapeutic agent to slow the progress of HCC.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Fructosediphosphates/pharmacology , Gene Expression Regulation/drug effects , Interleukin-8/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Hep G2 Cells , Humans , Inflammation/metabolism , Interleukin-8/genetics , Oxidative Stress/drug effects
12.
Stem Cell Res Ther ; 8(1): 36, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28202059

ABSTRACT

BACKGROUND: Heart failure represents an important public health issue due to its high costs and growing incidence worldwide. Evidence showing the regenerative potential of postmitotic heart tissue has suggested the existence of endogenous cardiac stem cells in adult hearts. Cardiosphere-derived cells (CDC) constitute a candidate pool of such cardiac stem cells. Previous studies using acute myocardial infarction (MI) models in rodents demonstrated an improvement in cardiac function after cell therapy with CDC. We evaluated the therapeutic potential of CDC 60 days after MI in a rat model. METHODS: CDC were obtained from human discarded myocardial tissue and rat hearts by enzymatic digestion with collagenase II. At 10-15 days after isolation, small, round, phase-bright cells (PBCs) appeared on top of the adherent fibroblast-like cells. The PBCs were collected and placed on a nonadherent plate for 2 days, where they formed cardiospheres which were then transferred to adherent plates, giving rise to CDC. These CDC were characterized by flow cytometry. Wistar rats were submitted to MI through permanent occlusion of the anterior descending coronary artery. After 60 days, they were immunosuppressed with cyclosporine A during 10 days. On the third day, infarcted animals were treated with 5 × 105 human CDC (hCDC) or placebo through intramyocardial injection guided by echocardiogram. Another group of animals was treated with rat CDC (rCDC) without immunosuppression. hCDC and rCDC were stably transduced with a viral construct expressing luciferase under control of a constitutive promoter. CDC were then used in a bioluminescence assay. Functional parameters were evaluated by echocardiogram 90 and 120 days after MI and by Langendorff at 120 days. RESULTS: CDC had a predominantly mesenchymal phenotype. Cell tracking by bioluminescence demonstrated over 85% decrease in signal at 5-7 days after cell therapy. Cardiac function evaluation by echocardiography showed no differences in ejection fraction, end-diastolic volume, or end-systolic volume between groups receiving human cells, rat cells, or placebo. Hemodynamic analyses and infarct area quantification confirmed that there was no improvement in cardiac remodeling after cell therapy with CDC. CONCLUSION: Our study challenges the effectiveness of CDC in post-ischemic heart failure.


Subject(s)
Coronary Occlusion/therapy , Immunocompromised Host , Myocardial Infarction/therapy , Spheroids, Cellular/transplantation , Animals , Coronary Occlusion/diagnostic imaging , Coronary Occlusion/immunology , Coronary Occlusion/physiopathology , Coronary Vessels/diagnostic imaging , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Cyclosporine/administration & dosage , Disease Models, Animal , Echocardiography , Heart Function Tests , Humans , Injections, Intralesional , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/immunology , Myocardial Infarction/physiopathology , Rats , Rats, Wistar , Spheroids, Cellular/cytology , Spheroids, Cellular/physiology , Stem Cells/cytology , Stem Cells/physiology , Treatment Failure
13.
J Hepatol ; 66(1): 86-94, 2017 01.
Article in English | MEDLINE | ID: mdl-27545498

ABSTRACT

BACKGROUND & AIMS: The transcription factor Krüppel-like factor 2 (KLF2), inducible by simvastatin, confers endothelial vasoprotection. Considering recent data suggesting activation of autophagy by statins, we aimed to: 1) characterize the relationship between autophagy and KLF2 in the endothelium, 2) assess this relationship in acute liver injury (cold ischemia/reperfusion) and 3) study the effects of modulating KLF2-autophagy in vitro and in vivo. METHODS: Autophagic flux, the vasoprotective KLF2 pathway, cell viability and microvascular function were assessed in endothelial cells and in various pre-clinical models of acute liver injury (cold storage and warm reperfusion). RESULTS: Positive feedback between autophagy and KLF2 was observed in the endothelium: KLF2 inducers, pharmacological (statins, resveratrol, GGTI-298), biomechanical (shear stress) or genetic (adenovirus containing KLF2), caused endothelial KLF2 overexpression through a Rac1-rab7-autophagy dependent mechanism, both in the specialized liver sinusoidal endothelial cells (LSEC) and in human umbilical vein endothelial cells. In turn, KLF2 induction promoted further activation of autophagy. Cold ischemia blunted autophagic flux. Upon reperfusion, LSEC stored in University of Wisconsin solution did not reactivate autophagy, which resulted in autophagosome accumulation probably due to impairment in autophagosome-lysosome fusion, ultimately leading to increased cell death and microvascular dysfunction. Simvastatin pretreatment maintained autophagy (through the upregulation of rab7), resulting in increased KLF2, improved cell viability, and ameliorated hepatic damage and microvascular function. CONCLUSIONS: We herein describe for the first time the complex autophagy-KLF2 relationship, modulating the phenotype and survival of the endothelium. These results help understanding the mechanisms of protection conferred by KLF2-inducers, such as simvastatin, in hepatic vascular disorders. LAY SUMMARY: Autophagy and the transcription factor KLF2 share a common activation pathway in the endothelium, being able to regulate each other. Statins maintain microvascular function through the inhibition of Rac1, which consequently liberates Rab7, activates autophagy and increments the expression of KLF2.


Subject(s)
Autophagy/physiology , Endothelium, Vascular , Liver Failure, Acute , Reperfusion Injury , Cell Survival , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kruppel-Like Transcription Factors/metabolism , Liver Failure, Acute/metabolism , Liver Failure, Acute/prevention & control , Microvessels/metabolism , Microvessels/physiopathology , Models, Biological , Protective Agents/pharmacology , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins , rac1 GTP-Binding Protein/metabolism
14.
Biomed Pharmacother ; 84: 1282-1290, 2016 12.
Article in English | MEDLINE | ID: mdl-27810785

ABSTRACT

Hepatocellular carcinoma is the most prevalent primary liver tumor and is among the top ten cancer that affect the world population. Its development is related, in most cases, to the existence of chronic liver injury, such as in cirrhosis. The knowledge about the correlation between chronic inflammation and cancer has driven new researches with anti-inflammatory agents that have potential for the development of antitumor drugs. Gallic acid is a phenolic acid found in many natural products and have shown anti-inflammatory, anti-tumor, anti-mutagenic and antioxidant actions. The purpose of this study was to investigate the effect of gallic acid on acute and chronic cell proliferation and inflammatory parameters of hepatocellular carcinoma cells (HepG2), as well as to investigate the mechanisms involved. Results showed that the gallic acid decreased the proliferation of HepG2 cells in a dose-dependent manner (Trypan blue exclusion assay), without causing necrosis (LDH assay). We observed a significant increase in the percentage of small and regular nuclei (Nuclear Morphometric Analysis assay), a significant induction of apoptosis by Annexin V-FITC and PI assay and no interference with the cell cycle using the FITC BrdU Flow Kit. We observed a significant reduction in the levels of IL-8 and increased levels of IL-10 and IL-12 (Cytometric Bead Array Human Inflammation Assay). Furthermore, gallic acid caused no cancer cells regrowth at a long term (Cumulative Population Doubling assay). According to these results, gallic acid showed a strong potential as an anti-tumor agent in hepatocellular carcinoma cells.


Subject(s)
Apoptosis/drug effects , Gallic Acid/pharmacology , Interleukin-8/metabolism , Biomarkers, Tumor/metabolism , Cell Cycle/drug effects , Cell Nucleus Shape/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Flow Cytometry , Gallic Acid/chemistry , Hep G2 Cells , Humans , Inflammation Mediators/metabolism , L-Lactate Dehydrogenase/metabolism
15.
Oncol Rep ; 36(5): 2647-2652, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27665945

ABSTRACT

Hepatocellular carcinoma is the most prevalent type of tumor among primary tumors affecting the liver. Rapamycin is currently used as a basis for chemotherapy in the treatment of cancers, including the liver. Because it shows several adverse effects, minimizing these effects without compromising efficacy is important. In this sense other drugs may be used concomitantly. One of these drugs is fructose-1,6-bisphosphate (FBP), which has shown therapeutic effect in various pathological situations, having antioxidant and anti-inflammatory proprieties. The objective of the present study was to evaluate the activity of rapamycin in combination with the FBP in HepG2 cell proliferation and the mechanisms involved. HepG2 cells were analyzed after 72 h of treatment with both drugs. Cell proliferation, cytotoxicity, cytokines, apoptosis, senescence, autophagy and oxidative stress were accessed. Ιt was demonstrated that the combination is more efficient than the single use of substances, because subtherapeutic doses of rapamycin, when associated to FBP become effective, reducing cell proliferation, through a significant increase in the production of tiobarbituric acid reactive substances (TBARS), suggesting that this might be the cause of death by apoptosis. According to these results, we believe that the association of both drugs may be a promising choice for the treatment of hepatocarcinoma.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Fructose-Bisphosphatase/administration & dosage , Liver Neoplasms/drug therapy , Sirolimus/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antioxidants/administration & dosage , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Free Radicals/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/pathology
16.
Toxicol In Vitro ; 30(1 Pt B): 309-17, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26475966

ABSTRACT

Apoptosis and NETosis of neutrophils are two major mechanisms of programmed cell death that differ in their morphological characteristics and effects on the immune system. Apoptosis can be delayed by the presence of pathogens or chemical components such as lipopolysaccharide (LPS). Neutrophils have other antimicrobial strategy, called neutrophil extracellular traps (NETs), which contributes to the elimination and control of the pathogen. NETosis is induced by infection, inflammation or trauma and represents an innate immune activation mechanism. The objective of this study was to evaluate the effect of gallic acid (GA) in the modulation of apoptosis and NETs release. The results show that GA decreased the anti-apoptotic effect of LPS, blocked the induction of NETs and prevented the formation of free radicals induced by LPS. These findings demonstrate that the GA is a novel therapeutic agent for decreasing the exacerbated response of the body against an infectious agent.


Subject(s)
Apoptosis/drug effects , Extracellular Traps/drug effects , Gallic Acid/pharmacology , Lipopolysaccharides/toxicity , Neutrophils/drug effects , Caspase 3/metabolism , Cytokines/metabolism , Humans
17.
Stem Cell Res ; 15(3): 445-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26413783

ABSTRACT

The human ihFib3.2 iPS cell line was generated from dermal fibroblasts obtained from a healthy donor. Lentiviral particles were produced with the polycistronic hSTEMCCA vector with Oct4, Sox2, cMyc and Klf4 as reprogramming factors.


Subject(s)
Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Cell Line , Cells, Cultured , Humans , Kruppel-Like Factor 4
18.
Inflamm Res ; 63(9): 719-28, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24888322

ABSTRACT

OBJECTIVE AND DESIGN: Mesenchymal stem cells (MSCs) are potent modulators of immune responses. Sepsis is the association of a systemic inflammatory response with an infection. The aim of this study was to test the ability of MSCs derived from adipose tissue, which have immunomodulatory effects, and to inhibit the septic process in an experimental model of mice. METHODS: Three experimental groups (male C57BL/6 mice) were formed for the test: control group, untreated septic group and septic group treated with MSCs (1 × 10(6) cells/animal). RESULTS: In the control group, there were no deaths; in the untreated septic group, the mortality rate was 100 % within 26 h; in the septic group treated with MSCs, the mortality rate reached 40 % within 26 h. The group treated with MSCs was able to reduce the markers of tissue damage in the liver and pancreas. The treated group had a reduction of inflammatory markers. Furthermore, the MSCs-treated group was able to inhibit the increase of apoptosis in splenocytes observed in the untreated septic group. CONCLUSIONS: Our data showed that MSCs ameliorated the immune response with decrease of inflammatory cytokines and increase anti-inflammatory IL-10; moreover, inhibited splenocytes apoptosis and, consequently, inhibited tissue damage during sepsis.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Sepsis/therapy , Spleen/cytology , Alanine Transaminase/blood , Amylases/blood , Animals , Apoptosis , Aspartate Aminotransferases/blood , Blood Glucose/analysis , Cells, Cultured , Cytokines/blood , Disease Models, Animal , Male , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Sepsis/blood , Sepsis/immunology , Transforming Growth Factor beta1/blood
19.
Cell Biol Int ; 38(4): 526-30, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24353036

ABSTRACT

(+)-Catechin is a type of catechin present in large amounts in açaí fruits and cocoa seeds. Besides its antioxidant and anti-inflammatory activities, little is known about its effects in the liver, especially during hepatic fibrosis. We report here the effects of (+)-catechin on hepatic stellate cells. (+)-Catechin induced quiescent phenotype in GRX cells, along with an increase in lipid droplets. Proliferator-activated receptor γ mRNA expression was upregulated, whereas type I collagen mRNA expression was downregulated. Pro-inflammatory cytokines were not influenced by (+)-catechin, whereas the levels of interleukin 10 were significantly increased. The data provide evidence that (+)-catechin can reduce hepatic stellate cell activation.


Subject(s)
Catechin/pharmacology , Hepatic Stellate Cells/drug effects , Animals , Cell Line , Collagen Type I/genetics , Collagen Type I/metabolism , Cytokines/metabolism , Down-Regulation/drug effects , Hepatic Stellate Cells/cytology , Interleukin-10/metabolism , Lipids/biosynthesis , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/metabolism , Stereoisomerism , Up-Regulation/drug effects
20.
Biochem Cell Biol ; 90(4): 575-84, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22574829

ABSTRACT

The phenolic compounds present in cocoa seeds have been studied regarding health benefits, such as antioxidant and anti-inflammatory activities. Fibrosis is a wound healing response that occurs in almost all patients with chronic liver injury. A large number of cytokines and soluble intercellular mediators are related to changes in the behavior and phenotype of the hepatic stellate cell (HSC) that develop a fibrogenic and contractile phenotype leading to the development of fibrosis. The objective of this study was to assess the catechin effect in GRX liver cells in activities such as cell growth and inflammation. The GRX cells treatment with catechin induced a significant decrease in cell growth. This mechanism does not occur by apoptosis or even by autophagy because there were no alterations in expression of caspase 3 and PARP (apoptosis), and LC3 (autophagy). The expression of p27 and p53 proteins, regulators of the cell cycle, showed increased expression, while COX-2 and IL-6 mRNA showed a significant decrease in expression. This study shows that catechin decreases cell growth in GRX cells and, probably, this decrease does not occur by apoptosis or autophagy but through an anti-inflammatory effect and cell cycle arrest. Catechin also significantly decreased the production of TGF-ß by GRX cells, showing a significant antifibrotic effect.


Subject(s)
Catechin/pharmacology , Hepatic Stellate Cells/drug effects , Autophagy , Cacao/chemistry , Caspase 3/metabolism , Cell Proliferation , Cells, Cultured , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Humans , Interleukin-6/metabolism , Liver/cytology , Liver/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...