Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
3.
Nat Commun ; 13(1): 5677, 2022 09 27.
Article in English | MEDLINE | ID: mdl-36167809

ABSTRACT

Fasting exerts beneficial effects in mice and humans, including protection from chemotherapy toxicity. To explore the involved mechanisms, we collect blood from humans and mice before and after 36 or 24 hours of fasting, respectively, and measure lipid composition of erythrocyte membranes, circulating micro RNAs (miRNAs), and RNA expression at peripheral blood mononuclear cells (PBMCs). Fasting coordinately affects the proportion of polyunsaturated versus saturated and monounsaturated fatty acids at the erythrocyte membrane; and reduces the expression of insulin signaling-related genes in PBMCs. When fasted for 24 hours before and 24 hours after administration of oxaliplatin or doxorubicin, mice show a strong protection from toxicity in several tissues. Erythrocyte membrane lipids and PBMC gene expression define two separate groups of individuals that accurately predict a differential protection from chemotherapy toxicity, with important clinical implications. Our results reveal a mechanism of fasting associated with lipid homeostasis, and provide biomarkers of fasting to predict fasting-mediated protection from chemotherapy toxicity.


Subject(s)
Fasting , MicroRNAs , Animals , Biomarkers , Doxorubicin/toxicity , Fasting/metabolism , Fatty Acids/metabolism , Fatty Acids, Monounsaturated , Homeostasis , Humans , Insulin , Leukocytes, Mononuclear/metabolism , Mice , Oxaliplatin
4.
Mar Drugs ; 19(9)2021 Sep 11.
Article in English | MEDLINE | ID: mdl-34564179

ABSTRACT

Phaeodactylum tricornutum is a rich source of fucoxanthin, a carotenoid with several health benefits. In the present study, high performance countercurrent chromatography (HPCCC) was used to isolate fucoxanthin from an extract of P. tricornutum. A multiple sequential injection HPCCC method was developed combining two elution modes (reverse phase and extrusion). The lower phase of a biphasic solvent system (n-heptane, ethyl acetate, ethanol and water, ratio 5/5/6/3, v/v/v/v) was used as the mobile phase, while the upper phase was the stationary phase. Ten consecutive sample injections (240 mg of extract each) were performed leading to the separation of 38 mg fucoxanthin with purity of 97% and a recovery of 98%. The process throughput was 0.189 g/h, while the efficiency per gram of fucoxanthin was 0.003 g/h. Environmental risk and general process evaluation factors were used for assessment of the developed separation method and compared with existing fucoxanthin liquid-liquid isolation methods. The isolated fucoxanthin retained its well-described ability to induce nuclear translocation of transcription factor FOXO3. Overall, the developed isolation method may represent a useful model to produce biologically active fucoxanthin from diatom biomass.


Subject(s)
Diatoms/chemistry , Xanthophylls/chemistry , Animals , Chromatography, High Pressure Liquid , Countercurrent Distribution
5.
Mol Biol Evol ; 38(8): 3415-3435, 2021 07 29.
Article in English | MEDLINE | ID: mdl-33871658

ABSTRACT

Aging and cancer are two interrelated processes, with aging being a major risk factor for the development of cancer. Parallel epigenetic alterations have been described for both, although differences, especially within the DNA hypomethylation scenario, have also been recently reported. Although many of these observations arise from the use of mouse models, there is a lack of systematic comparisons of human and mouse epigenetic patterns in the context of disease. However, such comparisons are significant as they allow to establish the extent to which some of the observed similarities or differences arise from pre-existing species-specific epigenetic traits. Here, we have used reduced representation bisulfite sequencing to profile the brain methylomes of young and old, tumoral and nontumoral brain samples from human and mouse. We first characterized the baseline epigenomic patterns of the species and subsequently focused on the DNA methylation alterations associated with cancer and aging. Next, we described the functional genomic and epigenomic context associated with the alterations, and finally, we integrated our data to study interspecies DNA methylation levels at orthologous CpG sites. Globally, we found considerable differences between the characteristics of DNA methylation alterations in cancer and aging in both species. Moreover, we describe robust evidence for the conservation of the specific cancer and aging epigenomic signatures in human and mouse. Our observations point toward the preservation of the functional consequences of these alterations at multiple levels of genomic regulation. Finally, our analyses reveal a role for the genomic context in explaining disease- and species-specific epigenetic traits.


Subject(s)
Aging/genetics , DNA Methylation , Epigenesis, Genetic , Epigenome , Neoplasms/genetics , Animals , Biological Evolution , CpG Islands , Humans , Mice , Species Specificity
7.
Aging (Albany NY) ; 12(12): 11337-11348, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32584785

ABSTRACT

Senescent cells accumulate with obesity in the white adipose tissue of mice and humans. These senescent cells enhance the pro-inflammatory environment that, with time, contributes to the onset of glucose intolerance and type 2 diabetes. Glucose intolerance in mouse models of obesity has been successfully reversed by the elimination of senescent cells with the senolytic compounds navitoclax or the combination of dasatinib and quercetin (D/Q). In this work, we generated obese mice by high-fat diet feeding, and treated them with five consecutive cycles of navitoclax or D/Q during 16 weeks. We observed an efficient reduction in the white adipose tissue of the senescence markers senescence-associated ß-galactosidase activity, Cdkn2a-p16 and Cdkn2a-p19 at the end of the 5 cycles. Mice treated with both navitoclax and D/Q showed an improvement of their insulin sensitivity and glucose tolerance during a short period of time (cycles 3 and 4), that disappeared at the fifth cycle. Also, these mice tended to increase the expression at their adipose tissue of the adipogenic genes Pparg and, Cebpa, as well as their plasma adiponectin levels. Together, our work shows that two different senolytic treatments, acting through independent pathways, are transiently effective in the treatment of obesity-induced metabolic disorders.


Subject(s)
Aniline Compounds/administration & dosage , Cellular Senescence/drug effects , Dasatinib/administration & dosage , Obesity/drug therapy , Quercetin/administration & dosage , Sulfonamides/administration & dosage , Adipogenesis/drug effects , Adiponectin/blood , Adiponectin/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cellular Senescence/physiology , Diet, High-Fat/adverse effects , Disease Models, Animal , Drug Administration Schedule , Drug Combinations , Glucose Intolerance/blood , Glucose Intolerance/drug therapy , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Humans , Insulin Resistance , Male , Mice , Mice, Obese , Obesity/blood , Obesity/etiology , Obesity/metabolism , PPAR gamma/metabolism
8.
Nat Commun ; 10(1): 4731, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31636264

ABSTRACT

Compounds with specific cytotoxic activity in senescent cells, or senolytics, support the causal involvement of senescence in aging and offer therapeutic interventions. Here we report the identification of Cardiac Glycosides (CGs) as a family of compounds with senolytic activity. CGs, by targeting the Na+/K+ATPase pump, cause a disbalanced electrochemical gradient within the cell causing depolarization and acidification. Senescent cells present a slightly depolarized plasma membrane and higher concentrations of H+, making them more susceptible to the action of CGs. These vulnerabilities can be exploited for therapeutic purposes as evidenced by the in vivo eradication of tumors xenografted in mice after treatment with the combination of a senogenic and a senolytic drug. The senolytic effect of CGs is also effective in the elimination of senescence-induced lung fibrosis. This experimental approach allows the identification of compounds with senolytic activity that could potentially be used to develop effective treatments against age-related diseases.


Subject(s)
Apoptosis/drug effects , Cardiac Glycosides/pharmacology , Cellular Senescence/drug effects , Chondrocytes/drug effects , Fibroblasts/drug effects , A549 Cells , Animals , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Breast Neoplasms , Cell Line, Tumor , Cell Membrane/drug effects , Digoxin/pharmacology , Female , Humans , Hydrogen-Ion Concentration/drug effects , Mice , Osteoarthritis , Ouabain/pharmacology , Proscillaridin/pharmacology , Pulmonary Fibrosis , Xenograft Model Antitumor Assays
9.
Methods Mol Biol ; 1890: 151-161, 2019.
Article in English | MEDLINE | ID: mdl-30414152

ABSTRACT

FOXO proteins are transcription factors with important roles in the regulation of the expression of genes involved in cell growth, proliferation, differentiation, and longevity. FOXO proteins are active in the nucleus but, upon post-translational modification they form a docking site for 14-3-3 proteins and are translocated to the cytoplasm where they are inactive.We make use of this regulatory mechanism of FOXO proteins to develop an image-based high-throughput screening platform to detect compounds that regulate FOXO3 subcellular localization. This system has proven a powerful tool to isolate inhibitors of proteins upstream of FOXO, such as PI3K inhibitors.


Subject(s)
Drug Discovery/methods , Forkhead Transcription Factors/genetics , High-Throughput Screening Assays , Transcriptional Activation/drug effects , Animals , Cell Line, Tumor , Data Analysis , Fluorescent Antibody Technique , Forkhead Transcription Factors/metabolism , Genes, Reporter , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Small Molecule Libraries
10.
EMBO Rep ; 19(9)2018 09.
Article in English | MEDLINE | ID: mdl-30021836

ABSTRACT

The NAD+-dependent deacetylase SIRT1 can be oncogenic or tumor suppressive depending on the tissue. Little is known about the role of SIRT1 in non-small cell lung carcinoma (NSCLC), one of the deadliest cancers, that is frequently associated with mutated K-RAS Therefore, we investigated the effect of SIRT1 on K-RAS-driven lung carcinogenesis. We report that SIRT1 protein levels are downregulated by oncogenic K-RAS in a MEK and PI3K-dependent manner in mouse embryo fibroblasts (MEFs), and in human lung adenocarcinoma cell lines. Furthermore, Sirt1 overexpression in mice delays the appearance of K-RasG12V-driven lung adenocarcinomas, reducing the number and size of carcinomas at the time of death and extending survival. Consistently, lower levels of SIRT1 are associated with worse prognosis in human NSCLCs. Mechanistically, analysis of mouse Sirt1-Tg pneumocytes, isolated shortly after K-RasG12V activation, reveals that Sirt1 overexpression alters pathways involved in tumor development: proliferation, apoptosis, or extracellular matrix organization. Our work demonstrates a tumor suppressive role of SIRT1 in the development of K-RAS-driven lung adenocarcinomas in mice and humans, suggesting that the SIRT1-K-RAS axis could be a therapeutic target for NSCLCs.


Subject(s)
Adenocarcinoma of Lung/metabolism , Carcinogenesis/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Sirtuin 1/metabolism , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Alveolar Epithelial Cells , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cells, Cultured , Down-Regulation , Fibroblasts/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Targeted Therapy , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Progression-Free Survival , Proto-Oncogene Proteins p21(ras)/genetics
11.
Genes Dev ; 23(10): 1177-82, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19451218

ABSTRACT

The INK4a/ARF tumor suppressor locus, a key executor of cellular senescence, is regulated by members of the Polycomb group (PcG) of transcriptional repressors. Here we show that signaling from oncogenic RAS overrides PcG-mediated repression of INK4a by activating the H3K27 demethylase JMJD3 and down-regulating the methyltransferase EZH2. In human fibroblasts, JMJD3 activates INK4a, but not ARF, and causes p16(INK4a)-dependent arrest. In mouse embryo fibroblasts, Jmjd3 activates both Ink4a and Arf and elicits a p53-dependent arrest, echoing the effects of RAS in this system. Our findings directly implicate JMJD3 in the regulation of INK4a/ARF during oncogene-induced senescence and suggest that JMJD3 has the capacity to act as a tumor suppressor.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Epigenesis, Genetic/genetics , Gene Expression Regulation , Oxidoreductases, N-Demethylating/metabolism , ras Proteins/metabolism , Animals , Cellular Senescence , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases , Mice , Signal Transduction
12.
Mutat Res ; 594(1-2): 78-85, 2006 Feb 22.
Article in English | MEDLINE | ID: mdl-16280139

ABSTRACT

Frequent chromosome 3 losses have been described in several tumors types, which strongly suggest the presence of one or several tumor suppressor genes. Recently, a novel candidate tumor suppressor gene termed Ris-1 (for Ras-induced senescence 1) has been identified at chromosomal position 3p21.3. Ris-1 has been proposed to participate in anti-tumor responses that resemble cellular senescence and that are elicited by oncogenes such as Ras. To analyze the role of Ris-1 as a putative tumor suppressor gene in human breast cancer, we have performed a real-time quantitative analysis of its mRNA expression in 60 patients. Moreover, we carried out a first approach to evaluate the most common inactivation mechanism that can affect expression levels of tumor suppressor genes (mutation, promoter hypermethylation and allelic losses). Furthermore, a correlation study between expression as well as inactivating mechanisms of Ris-1 and several clinico-pathological parameters of the tumors was designed, with the objective of appraising the prognostic value of Ris-1 status. Decreased expression of Ris-1 was observed in 23% of the cases and overexpressed Ris-1 was detected in 15% of the primary breast tumors. Our data showed high frequency of LOH (30%) at one of the markers used. Nevertheless, a polymorphism related with the expression levels was described. Statistically significant correlations were found between decreased Ris-1 expression and negative progesterone receptors, as well as between overexpressing Ris-1 tumors and high histological grade. Despite all these data, we conclude that the suggested role of Ris-1 as tumor suppressor gene is not evident, at least in breast cancer. Future and larger series studies in different tumor types are necessary to clarify Ris-1 function in human cancer.


Subject(s)
Breast Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Alleles , Breast Neoplasms/physiopathology , DNA Methylation , Epigenesis, Genetic , Female , Genotype , Humans , Membrane Proteins , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Promoter Regions, Genetic , Prospective Studies , Tumor Suppressor Proteins/biosynthesis
13.
Nature ; 436(7051): 642, 2005 Aug 04.
Article in English | MEDLINE | ID: mdl-16079833

ABSTRACT

Oncogene-induced senescence is a cellular response that may be crucial for protection against cancer development, but its investigation has so far been restricted to cultured cells that have been manipulated to overexpress an oncogene. Here we analyse tumours initiated by an endogenous oncogene, ras, and show that senescent cells exist in premalignant tumours but not in malignant ones. Senescence is therefore a defining feature of premalignant tumours that could prove valuable in the diagnosis and prognosis of cancer.


Subject(s)
Cellular Senescence , Lung Neoplasms/pathology , Precancerous Conditions/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Alleles , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Genes, ras/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
14.
Cancer Cell ; 4(2): 111-20, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12957286

ABSTRACT

We have targeted a K-ras allele in mouse embryonic stem (ES) cells to express a K-Ras(V12) oncoprotein along with a marker protein (beta-geo) from a single bicistronic transcript. Expression of this oncogenic allele requires removal of a knocked in STOP transcriptional cassette by Cre recombinase. Primary mouse embryonic fibroblasts expressing this K-ras(V12) allele do not undergo proliferative senescence and proliferate as immortal cells. In mice, expression of K-ras(V12) throughout the body fails to induce unscheduled proliferation or other growth abnormalities for up to eight months. Only a percentage of K-ras(V12)-expressing lung bronchiolo-alveolar cells undergo malignant transformation leading to the formation of multiple adenomas and adenocarcinomas. These results indicate that neoplastic growth induced by an endogenous K-ras oncogene depends upon cellular context.


Subject(s)
Cell Transformation, Neoplastic , Fibroblasts/pathology , Genes, ras/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases , Animals , Cell Division , Cell Line, Transformed , Cellular Senescence , Chromosome Aberrations , Cyclin-Dependent Kinase Inhibitor p16 , Fibroblasts/metabolism , Gene Targeting , Genetic Vectors/genetics , MAP Kinase Signaling System , Mice , Mice, Transgenic , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/pathology , Survival Rate , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Exp Cell Res ; 273(2): 127-37, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11822868

ABSTRACT

Normal cells display protective responses against oncogenes. Notably, oncogenic Ras triggers an irreversible proliferation arrest that is reminiscent of replicative senescence and that is considered a relevant tumor-suppressor mechanism. Here, we have used microarrayed filters to identify genes specifically upregulated in Ras-senescent human fibroblasts. Among the initial set of genes selected from the microarrays, we found the cell-cycle inhibitor p21(Cip1/Waf1), thus validating the potency of the screening to identify markers and mediators of Ras-senescence. A group of six genes, formed by those more highly upregulated during Ras-senescence, was analyzed in further detail to evaluate their specificity. In particular, we examined their expression in cells overexpressing Ras but rendered resistant to Ras-senescence by the viral oncoprotein E1a; also, we have studied their expression during replicative senescence, organismal aging, H(2)O(2)-induced senescence, and DNA damage. In this manner, we have identified a novel gene, RIS1 (for Ras-induced senescence 1), which is not upregulated in association to any of the above-mentioned processes, but exclusively during Ras-senescence. Furthermore, RIS1 is also upregulated by the transcriptional factor Ets2, which is a known mediator of Ras-induced senescence. Interestingly, RIS1 is located at chromosomal position 3p21.3 and, more specifically, it is included in a short segment of just 1 Mb previously defined by other investigators for its tumor-suppressor activity. In summary, we report the identification of a novel gene, RIS1, as a highly specific marker of Ras-induced senescence and a candidate tumor-suppressor gene.


Subject(s)
DNA-Binding Proteins , Genes, Tumor Suppressor , Repressor Proteins , Transcription Factors , Transcriptional Activation , Tumor Suppressor Proteins/genetics , ras Proteins/metabolism , Adenovirus E1A Proteins/metabolism , Adenovirus E1A Proteins/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Cell Division , Cell Line , Cellular Senescence , Child , DNA Damage , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Hydrogen Peroxide/pharmacology , Membrane Proteins , Middle Aged , Proto-Oncogene Protein c-ets-2 , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Up-Regulation , ras Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...