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.
J Exp Clin Cancer Res ; 40(1): 248, 2021 Aug 07.
Article in English | MEDLINE | ID: mdl-34364401

ABSTRACT

BACKGROUND: The identification of novel targets is of paramount importance to develop more effective drugs and improve the treatment of non-small cell lung cancer (NSCLC), the leading cause of cancer-related deaths worldwide. Since cells alter their metabolic rewiring during tumorigenesis and along cancer progression, targeting key metabolic players and metabolism-associated proteins represents a valuable approach with a high therapeutic potential. Metabolic fitness relies on the functionality of heat shock proteins (HSPs), molecular chaperones that facilitate the correct folding of metabolism enzymes and their assembly in macromolecular structures. METHODS: Gene fitness was determined by bioinformatics analysis from available datasets from genetic screenings. HSPD1 expression was evaluated by immunohistochemistry from formalin-fixed paraffin-embedded tissues from NSCLC patients. Real-time proliferation assays with and without cytotoxicity reagents, colony formation assays and cell cycle analyses were used to monitor growth and drug sensitivity of different NSCLC cells in vitro. In vivo growth was monitored with subcutaneous injections in immune-deficient mice. Cell metabolic activity was analyzed through extracellular metabolic flux analysis. Specific knockouts were introduced by CRISPR/Cas9. RESULTS: We show heat shock protein family D member 1 (HSPD1 or HSP60) as a survival gene ubiquitously expressed in NSCLC and associated with poor patients' prognosis. HSPD1 knockdown or its chemical disruption by the small molecule KHS101 induces a drastic breakdown of oxidative phosphorylation, and suppresses cell proliferation both in vitro and in vivo. By combining drug profiling with transcriptomics and through a whole-genome CRISPR/Cas9 screen, we demonstrate that HSPD1-targeted anti-cancer effects are dependent on oxidative phosphorylation and validated molecular determinants of KHS101 sensitivity, in particular, the creatine-transporter SLC6A8 and the subunit of the cytochrome c oxidase complex COX5B. CONCLUSIONS: These results highlight mitochondrial metabolism as an attractive target and HSPD1 as a potential theranostic marker for developing therapies to combat NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Chaperonin 60/metabolism , Lung Neoplasms/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Disease Models, Animal , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , Survival Analysis
2.
Oncogene ; 40(12): 2309-2322, 2021 03.
Article in English | MEDLINE | ID: mdl-33654197

ABSTRACT

Since their discovery, microRNAs (miRNAs) have been widely studied in almost every aspect of biology and medicine, leading to the identification of important gene regulation circuits and cellular mechanisms. However, investigations are generally focused on the analysis of their downstream targets and biological functions in overexpression and knockdown approaches, while miRNAs endogenous levels and activity remain poorly understood. Here, we used the cellular plasticity-regulating process of epithelial-to-mesenchymal transition (EMT) as a model to show the efficacy of a fluorescent sensor to separate cells with distinct EMT signatures, based on miR-200b/c activity. The system was further combined with a CRISPR-Cas9 screening platform to unbiasedly identify miR-200b/c upstream regulating genes. The sensor allows to infer miRNAs fundamental biological properties, as profiling of sorted cells indicated miR-200b/c as a molecular switch between EMT differentiation and proliferation, and suggested a role for metabolic enzymes in miR-200/EMT regulation. Analysis of miRNAs endogenous levels and activity for in vitro and in vivo applications could lead to a better understanding of their biological role in physiology and disease.


Subject(s)
Cell Plasticity/genetics , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , HCT116 Cells , Humans
3.
Mater Sci Eng C Mater Biol Appl ; 111: 110832, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32279802

ABSTRACT

The physico-chemical characteristics of the extracellular matrix (ECM) cause mechanical cues that could elicit responses in the survival rate of cortical neuronal cells. Efficient neurite outgrowth in vitro, is critical for successful cultivation of cortical neuronal cells and the potential for attempts at regeneration of the central nervous system (CNS) in vivo. Relatively soft and hydrophilic, microbially synthesized aromatic polyester, polyhydroxyphenylvalerate (PHPV) was blended 50:50 with the stiff and hydrophobic polycaprolactone (PCL) and electrospun in microfibers for use in a 3D (CellCrown™) configuration and in a 2D coverslip coated configuration. This blend allows a 2.3-fold increase in the life-span of human induced pluripotent stem derived cortical neuronal cells (hiPS) compared to pure PCL fibers. HiPS-derived cortical neuronal cells grown on PHPV/PCL fibers show a 3.8-fold higher cumulative neurite elaboration compared to neurites grown on PCL fibers only. 96% of cortical neuronal cells die after 8 days of growth when plated on PCL fibers alone while >83% and 55% are alive on PHPV/PCL fibers on day 8 and day 17, respectively. An increased migration rate of cortical neuronal cells is also promoted by the blend compared to the PCL fibers alone. The critical survival rate improvement of hiPS derived cortical neuronal cells on PHPV/PCL blend holds promise in using these biocompatible nanofibers as implantable materials for regenerative purposes of an active cortical neuronal population after full maturation in vitro.


Subject(s)
Cerebral Cortex/cytology , Mechanotransduction, Cellular , Nanofibers/chemistry , Neurons/cytology , Polyesters/pharmacology , Caspase 3/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Mechanotransduction, Cellular/drug effects , Neurites/drug effects , Neurites/metabolism , Regression Analysis , Stress, Mechanical , Temperature
4.
Mol Metab ; 35: 100962, 2020 05.
Article in English | MEDLINE | ID: mdl-32244187

ABSTRACT

BACKGROUND: Nucleotide metabolism is a critical pathway that generates purine and pyrimidine molecules for DNA replication, RNA synthesis, and cellular bioenergetics. Increased nucleotide metabolism supports uncontrolled growth of tumors and is a hallmark of cancer. Agents inhibiting synthesis and incorporation of nucleotides in DNA are widely used as chemotherapeutics to reduce tumor growth, cause DNA damage, and induce cell death. Thus, the research on nucleotide metabolism in cancer is primarily focused on its role in cell proliferation. However, in addition to proliferation, the role of purine molecules is established as ligands for purinergic signals. However, so far, the role of the pyrimidines has not been discussed beyond cell growth. SCOPE OF THE REVIEW: In this review we present the key evidence from recent pivotal studies supporting the notion of a non-proliferative role for pyrimidine metabolism (PyM) in cancer, with a special focus on its effect on differentiation in cancers from different origins. MAJOR CONCLUSION: In leukemic cells, the pyrimidine catabolism induces terminal differentiation toward monocytic lineage to check the aberrant cell proliferation, whereas in some solid tumors (e.g., triple negative breast cancer and hepatocellular carcinoma), catalytic degradation of pyrimidines maintains the mesenchymal-like state driven by epithelial-to-mesenchymal transition (EMT). This review further broadens this concept to understand the effect of PyM on metastasis and, ultimately, delivers a rationale to investigate the involvement of the pyrimidine molecules as oncometabolites. Overall, understanding the non-proliferative role of PyM in cancer will lead to improvement of the existing antimetabolites and to development of new therapeutic options.


Subject(s)
Cell Proliferation , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Pyrimidines/metabolism , Animals , Cell Differentiation , Disease Progression , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology
5.
Cell Death Differ ; 26(11): 2223-2236, 2019 11.
Article in English | MEDLINE | ID: mdl-30737477

ABSTRACT

Cancer cells frequently boost nucleotide metabolism (NM) to support their increased proliferation, but the consequences of elevated NM on tumor de-differentiation are mostly unexplored. Here, we identified a role for thymidylate synthase (TS), a NM enzyme and established drug target, in cancer cell de-differentiation and investigated its clinical significance in breast cancer (BC). In vitro, TS knockdown increased the population of CD24+ differentiated cells, and attenuated migration and sphere-formation. RNA-seq profiling indicated repression of epithelial-to-mesenchymal transition (EMT) signature genes upon TS knockdown, and TS-deficient cells showed an increased ability to invade and metastasize in vivo, consistent with the occurrence of a partial EMT phenotype. Mechanistically, TS enzymatic activity was found essential for maintenance of the EMT/stem-like state by fueling a dihydropyrimidine dehydrogenase-dependent pyrimidine catabolism. In patient tissues, TS levels were found significantly higher in poorly differentiated and in triple negative BC, and strongly correlated with worse prognosis. The present study provides the rationale to study in-depth the role of NM at the crossroads of proliferation and differentiation, and depicts new avenues for the design of novel drug combinations for the treatment of BC.


Subject(s)
Cell Dedifferentiation/physiology , Thymidylate Synthase/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , CD24 Antigen/metabolism , Cell Movement , Cell Proliferation/physiology , Dihydrouracil Dehydrogenase (NADP)/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Prognosis , Pyrimidines/metabolism , Spheroids, Cellular , Thymidylate Synthase/genetics , Tumor Cells, Cultured
6.
Cancer Res ; 78(7): 1604-1618, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29343522

ABSTRACT

Cancer cells alter their metabolism to support their malignant properties. In this study, we report that the glucose-transforming polyol pathway (PP) gene aldo-keto-reductase-1-member-B1 (AKR1B1) strongly correlates with epithelial-to-mesenchymal transition (EMT). This association was confirmed in samples from lung cancer patients and from an EMT-driven colon cancer mouse model with p53 deletion. In vitro, mesenchymal-like cancer cells showed increased AKR1B1 levels, and AKR1B1 knockdown was sufficient to revert EMT. An equivalent level of EMT suppression was measured by targeting the downstream enzyme sorbitol-dehydrogenase (SORD), further pointing at the involvement of the PP. Comparative RNA sequencing confirmed a profound alteration of EMT in PP-deficient cells, revealing a strong repression of TGFß signature genes. Excess glucose was found to promote EMT through autocrine TGFß stimulation, while PP-deficient cells were refractory to glucose-induced EMT. These data show that PP represents a molecular link between glucose metabolism, cancer differentiation, and aggressiveness, and may serve as a novel therapeutic target.Significance: A glucose-transforming pathway in TGFß-driven epithelial-to-mesenchymal transition provides novel mechanistic insights into the metabolic control of cancer differentiation. Cancer Res; 78(7); 1604-18. ©2018 AACR.


Subject(s)
Aldehyde Reductase/genetics , Colonic Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , L-Iditol 2-Dehydrogenase/genetics , Lung Neoplasms/pathology , A549 Cells , Animals , Cell Line, Tumor , Glucose/metabolism , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , MCF-7 Cells , Mice , RNA Interference , RNA, Small Interfering/genetics , Transforming Growth Factor beta/metabolism
7.
J Pathol ; 242(2): 221-233, 2017 06.
Article in English | MEDLINE | ID: mdl-28337746

ABSTRACT

Thymidylate synthase (TS) is a fundamental enzyme of nucleotide metabolism and one of the oldest anti-cancer targets. Beginning from the analysis of gene array data from the NCI-60 panel of cancer cell lines, we identified a significant correlation at both gene and protein level between TS and the markers of epithelial-to-mesenchymal transition (EMT), a developmental process that allows cancer cells to acquire features of aggressiveness, like motility and chemoresistance. TS levels were found to be significantly augmented in mesenchymal-like compared to epithelial-like cancer cells, to be regulated by EMT induction, and to negatively correlate with micro-RNAs (miRNAs) usually expressed in epithelial-like cells and known to actively suppress EMT. Transfection of EMT-suppressing miRNAs reduced TS levels, and a specific role for miR-375 in targeting the TS 3'-untranslated region was identified. A particularly relevant association was found between TS and the powerful EMT driver ZEB1, the shRNA-mediated knockdown of which up-regulated miR-375 and reduced TS cellular levels. The TS-ZEB1 association was confirmed in clinical specimens from lung tumours and in a genetic mouse model of pancreatic cancer with ZEB1 deletion. Interestingly, TS itself appeared to have a regulatory role in EMT in cancer cells, as TS knockdown could directly reduce the EMT phenotype, the migratory ability of cells, the expression of stem-like markers, and chemoresistance. Taken together, these data indicate that the TS enzyme is functionally linked with EMT and cancer differentiation, with several potential translational implications. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Epithelial-Mesenchymal Transition , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Thymidylate Synthase/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Adult , Aged , Aged, 80 and over , Animals , Base Sequence , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , RNA, Small Interfering/genetics , Thymidylate Synthase/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...