Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol In Vitro ; 68: 104927, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32634469

ABSTRACT

Breast cancer is a complex disease and encompassing different types of tumor. Although advances in understanding of the molecular bases of breast cancer biology, the therapeutic proposals available still are not effective. In this scenario, the present study aimed to evaluate the mechanisms associated to antitumor activity of 7-Epiclusianone (7-Epi), a tetraprenylated benzophenone, on luminal A (MCF-7) and claudin-low (Hs 578T) breast cancer cell lines. We found that 7-Epi efficiently inhibited cell proliferation and migration of these cells; however MCF-7 was slightly more responsive than Hs 578T. Cell cycle analysis showed accumulation of cells at G0/G1 phase with drastic reduction of S population in treated cultures. This effect was associated to downregulation of CDKN1A (p21) and cyclin E in both cell lines. In addition, 7-Epi reduced cyclin D1 and p-ERK expression levels in MCF-7 cell line. Cytotoxic effect of 7-Epi on breast cancer cell lines was associated to its ability to increase BAX/BCL-2 ratio. In conclusion, our findings showed that 7-Epi is a promising antitumor agent against breast cancer by modulating critical regulators of the cell cycle and apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzophenones/pharmacology , Benzoquinones/pharmacology , Breast Neoplasms/drug therapy , Apoptosis/drug effects , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Humans , Proto-Oncogene Proteins c-bcl-2/genetics
2.
Hepatology ; 72(4): 1412-1429, 2020 10.
Article in English | MEDLINE | ID: mdl-32516515

ABSTRACT

BACKGROUND AND AIMS: Telomere attrition is a major risk factor for end-stage liver disease. Due to a lack of adequate models and intrinsic difficulties in studying telomerase in physiologically relevant cells, the molecular mechanisms responsible for liver disease in patients with telomere syndromes remain elusive. To circumvent that, we used genome editing to generate isogenic human embryonic stem cells (hESCs) harboring clinically relevant mutations in telomerase and subjected them to an in vitro, stage-specific hepatocyte differentiation protocol that resembles hepatocyte development in vivo. APPROACH AND RESULTS: Using this platform, we observed that while telomerase is highly expressed in hESCs, it is quickly silenced, specifically due to telomerase reverse transcriptase component (TERT) down-regulation, immediately after endoderm differentiation and completely absent in in vitro-derived hepatocytes, similar to what is observed in human primary hepatocytes. While endoderm derivation is not impacted by telomere shortening, progressive telomere dysfunction impaired hepatic endoderm formation. Consequently, hepatocyte derivation, as measured by expression of specific hepatic markers as well by albumin expression and secretion, is severely compromised in telomerase mutant cells with short telomeres. Interestingly, this phenotype was not caused by cell death induction or senescence. Rather, telomere shortening prevents the up-regulation and activation of human hepatocyte nuclear factor 4 alpha (HNF4α) in a p53-dependent manner. Both reactivation of telomerase and silencing of p53 rescued hepatocyte formation in telomerase mutants. Likewise, the conditional expression (doxycycline-controlled) of HNF4α, even in cells that retained short telomeres, accrued DNA damage, and exhibited p53 stabilization, successfully restored hepatocyte formation from hESCS. CONCLUSIONS: Our data show that telomere dysfunction acts as a major regulator of HNF4α during hepatocyte development, pointing to a target in the treatment of liver disease in telomere-syndrome patients.


Subject(s)
Hepatocyte Nuclear Factor 4/physiology , Hepatocytes/physiology , Telomere/physiology , Tumor Suppressor Protein p53/physiology , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells , Hepatocytes/cytology , Humans , Telomerase/genetics
3.
Cell Biol Int ; 42(1): 95-104, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28906033

ABSTRACT

Clinical data published in recent years have demonstrated positive effects of collagen hydrolysate (CH) on skin aging clinical signs. CH use as food supplement has a long history; however, few studies have addressed the underlying purpose of CH on the cellular and molecular biology of skin cells that could elucidate clinical improvement findings. Wide diversity of characteristics has been reported for dermal fibroblasts derived from different body sites and it is unknown whether collagen peptides could modulate differently cells from chronological aged and photoaged skin areas. This study investigated the influence of CH on the extracellular matrix metabolism and proliferation of human dermal fibroblasts (HDFs) derived from chronological aged (sun-protected) and photoaged (sun-exposed) body sites. CH treatment did not affect cellular proliferation of either cell cultures, but notably modulated cell metabolism in monolayer model, increasing the content of dermal matrix precursor and main protein, procollagen I and collagen I, respectively. These effects were confirmed in the human dermal equivalent model. The increase in collagen content in the cultures was attributed to stimulation of biosynthesis and decreased collagen I metabolism through inhibition of metalloproteinase activity (MMP) 1 and 2. Modulation of CH in dermal metabolism did not differ between cells derived from sun-protected and sun-exposed areas, although lower concentrations of CH seemed to be enough to stimulate sun-exposed-derived HDFs, suggesting more pronounced effect in these cells. This study contributes to understanding the biological effects of CH on skin cells and viability of its use as a functional ingredient in food supplements.


Subject(s)
Collagen/metabolism , Dermis/metabolism , Extracellular Matrix/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/metabolism , Fibroblasts/metabolism , Humans , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 2/metabolism , Peptides/metabolism , Skin/cytology , Skin Aging/physiology , Ultraviolet Rays/adverse effects
4.
Stem Cell Reports ; 9(2): 409-418, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28757166

ABSTRACT

Dyskeratosis congenita (DC) is a bone marrow failure syndrome associated with telomere dysfunction. The progression and molecular determinants of hematopoietic failure in DC remain poorly understood. Here, we use the directed differentiation of human embryonic stem cells harboring clinically relevant mutations in telomerase to understand the consequences of DC-associated mutations on the primitive and definitive hematopoietic programs. Interestingly, telomere shortening does not broadly impair hematopoiesis, as primitive hematopoiesis is not impaired in DC cells. In contrast, while phenotypic definitive hemogenic endothelium is specified, the endothelial-to-hematopoietic transition is impaired in cells with shortened telomeres. This failure is caused by DNA damage accrual and is mediated by p53 stabilization. These observations indicate that detrimental effects of telomere shortening in the hematopoietic system are specific to the definitive hematopoietic lineages. This work illustrates how telomere dysfunction impairs hematopoietic development and creates a robust platform for therapeutic discovery for treatment of DC patients.


Subject(s)
Dyskeratosis Congenita/blood , Dyskeratosis Congenita/genetics , Hematopoiesis/genetics , Tumor Suppressor Protein p53/genetics , Anemia, Aplastic/blood , Anemia, Aplastic/etiology , Anemia, Aplastic/pathology , Biomarkers , Bone Marrow/pathology , Bone Marrow Diseases/blood , Bone Marrow Diseases/etiology , Bone Marrow Diseases/pathology , Bone Marrow Failure Disorders , Cell Differentiation/genetics , DNA Damage , DNA Mutational Analysis , Dyskeratosis Congenita/pathology , Embryonic Stem Cells/metabolism , Gene Knockout Techniques , Gene Targeting , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hemoglobinuria, Paroxysmal/blood , Hemoglobinuria, Paroxysmal/etiology , Hemoglobinuria, Paroxysmal/pathology , Histones/metabolism , Humans , Immunophenotyping , Models, Biological , Mutation , Phenotype , Telomere , Telomere Homeostasis/genetics , Tumor Suppressor Protein p53/metabolism
5.
J Exp Clin Cancer Res ; 33: 37, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24775603

ABSTRACT

Some cancers like melanoma and pancreatic and ovarian cancers, for example, commonly display resistance to chemotherapy, and this is the major obstacle to a better prognosis of patients. Frequently, literature presents studies in monolayer cell cultures, 3D cell cultures or in vivo studies, but rarely the same work compares results of drug resistance in different models. Several of these works are presented in this review and show that usually cells in 3D culture are more resistant to drugs than monolayer cultured cells due to different mechanisms. Searching for new strategies to sensitize different tumors to chemotherapy, many methods have been studied to understand the mechanisms whereby cancer cells acquire drug resistance. These methods have been strongly advanced along the years and therapies using different drugs have been increasingly proposed to induce cell death in resistant cells of different cancers. Recently, cancer stem cells (CSCs) have been extensively studied because they would be the only cells capable of sustaining tumorigenesis. It is believed that the resistance of CSCs to currently used chemotherapeutics is a major contributing factor in cancer recurrence and later metastasis development. This review aims to appraise the experimental progress in the study of acquired drug resistance of cancer cells in different models as well as to understand the role of CSCs as the major contributing factor in cancer recurrence and metastasis development, describing how CSCs can be identified and isolated.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Drug Resistance, Multiple , Drug Screening Assays, Antitumor , Heat-Shock Proteins/metabolism , Humans , Neoplasm Recurrence, Local/prevention & control , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/physiology , Tumor Cells, Cultured
6.
J Exp Clin Cancer Res ; 32: 31, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23701745

ABSTRACT

Anticancer activities of cinnamic acid derivatives include induction of apoptosis by irreversible DNA damage leading to cell death. The present work aimed to compare the cytotoxic and genotoxic potential of cinnamic acid in human melanoma cell line (HT-144) and human melanocyte cell line derived from blue nevus (NGM). Viability assay showed that the IC50 for HT-144 cells was 2.4 mM, while NGM cells were more resistant to the treatment. The growth inhibition was probably associated with DNA damage leading to DNA synthesis inhibition, as shown by BrdU incorporation assay, induction of nuclear aberrations and then apoptosis. The frequency of cell death caused by cinnamic acid was higher in HT-144 cells. Activated-caspase 3 staining showed apoptosis after 24 hours of treatment with cinnamic acid 3.2 mM in HT-144 cells, but not in NGM. We observed microtubules disorganization after cinnamic acid exposure, but this event and cell death seem to be independent according to M30 and tubulin labeling. The frequency of micronucleated HT-144 cells was higher after treatment with cinnamic acid (0.4 and 3.2 mM) when compared to the controls. Cinnamic acid 3.2 mM also increased the frequency of micronucleated NGM cells indicating genotoxic activity of the compound, but the effects were milder. Binucleation and multinucleation counting showed similar results. We conclude that cinnamic acid has effective antiproliferative activity against melanoma cells. However, the increased frequency of micronucleation in NGM cells warrants the possibility of genotoxicity and needs further investigation.


Subject(s)
Apoptosis/drug effects , Cinnamates/pharmacology , Melanoma/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Survival/drug effects , Chromosome Aberrations/drug effects , Cinnamates/toxicity , Cytoskeleton/metabolism , Humans , Microtubules/metabolism , Phosphorylation/drug effects , Tubulin/metabolism , Tumor Suppressor Protein p53/metabolism
7.
Cancer Cell Int ; 13(1): 38, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23631593

ABSTRACT

BACKGROUND: Lung cancer often exhibits molecular changes, such as the overexpression of the ErbB1 gene. ErbB1 encodes epidermal growth factor receptor (EGFR), a tyrosine kinase receptor, involved mainly in cell proliferation and survival. EGFR overexpression has been associated with more aggressive disease, poor prognosis, low survival rate and low response to therapy. ErbB1 amplification and mutation are associated with tumor development and are implicated in ineffective treatment. The aim of the present study was to investigate whether the ErbB1 copy number affects EGFR expression, cell proliferation or cell migration by comparing two different cell lines. METHODS: The copies of ErbB1 gene was evaluated by FISH. Immunofluorescence and Western blotting were performed to determine location and expression of proteins mentioned in the present study. Proliferation was studied by flow cytometry and cell migration by wound healing assay and time lapse. RESULTS: We investigated the activation and function of EGFR in the A549 and HK2 lung cancer cell lines, which contain 3 and 6 copies of ErbB1, respectively. The expression of EGFR was lower in the HK2 cell line. EGFR was activated after stimulation with EGF in both cell lines, but this activation did not promote differences in cellular proliferation when compared to control cells. Inhibiting EGFR with AG1478 did not modify cellular proliferation, confirming previous data. However, we observed morphological alterations, changes in microfilament organization and increased cell migration upon EGF stimulation. However, these effects did not seem to be consequence of an epithelial-mesenchymal transition. CONCLUSION: EGFR expression did not appear to be associated to the ErbB1 gene copy number, and neither of these aspects appeared to affect cell proliferation. However, EGFR activation by EGF resulted in cell migration stimulation in both cell lines.

SELECTION OF CITATIONS
SEARCH DETAIL
...