Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 9549, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31267022

ABSTRACT

The existence of radio- and chemotherapy-surviving cancer stem cells is currently believed to explain the inefficacy of anti-glioblastoma (GBM) therapies. The aim of this study was to determine if a therapeutic strategy specifically targeting GBM stem cells (GSC) would completely eradicate a GBM tumor. In both the in vitro and the in vivo models, ganciclovir therapy targeting proliferating GSC promotes the survival of a quiescent, stem-like cell pool capable of reproducing the tumor upon release of the therapeutic pressure. Images of small niches of therapy-surviving tumor cells show organized networks of vascular-like structures formed by tumor cells expressing CD133 or OCT4/SOX2. These results prompted the investigation of tumor cells differentiated to endothelial and pericytic lineages as a potential reservoir of tumor-initiating capacity. Isolated tumor cells with pericyte and endothelial cell lineage characteristics, grown under tumorsphere forming conditions and were able to reproduce tumors after implantation in mice.


Subject(s)
AC133 Antigen/genetics , Glioma/genetics , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , SOXB1 Transcription Factors/genetics , AC133 Antigen/metabolism , Animals , Biomarkers , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Genes, Reporter , Glioma/drug therapy , Glioma/pathology , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism
2.
Mol Ther Oncolytics ; 11: 39-51, 2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30364660

ABSTRACT

A preclinical model of glioblastoma (GB) bystander cell therapy using human adipose mesenchymal stromal cells (hAMSCs) is used to address the issues of cell availability, quality, and feasibility of tumor cure. We show that a fast proliferating variety of hAMSCs expressing thymidine kinase (TK) has therapeutic capacity equivalent to that of TK-expressing hAMSCs and can be used in a multiple-inoculation procedure to reduce GB tumors to a chronically inhibited state. We also show that up to 25% of unmodified hAMSCs can be tolerated in the therapeutic procedure without reducing efficacy. Moreover, mimicking a clinical situation, tumor debulking previous to cell therapy inhibits GB tumor growth. To understand these striking results at a cellular level, we used a bioluminescence imaging strategy and showed that tumor-implanted therapeutic cells do not proliferate, are unaffected by GCV, and spontaneously decrease to a stable level. Moreover, using the CLARITY procedure for tridimensional visualization of fluorescent cells in transparent brains, we find therapeutic cells forming vascular-like structures that often associate with tumor cells. In vitro experiments show that therapeutic cells exposed to GCV produce cytotoxic extracellular vesicles and suggest that a similar mechanism may be responsible for the in vivo therapeutic effectiveness of TK-expressing hAMSCs.

3.
Adv Healthc Mater ; 7(17): e1800335, 2018 09.
Article in English | MEDLINE | ID: mdl-29923337

ABSTRACT

The encapsulation of mRNA in nanosystems as gene vaccines for immunotherapy purposes has experienced an exponential increase in recent years. Despite the many advantages envisaged within these approaches, their application in clinical treatments is still limited due to safety issues. These issues can be attributed, in part, to liver accumulation of most of the designed nanosystems and to the inability to transfect immune cells after an intravenous administration. In this context, this study takes advantage of the known versatile properties of the oligopeptide end-modified poly (ß-amino esters) (OM-PBAEs) to complex mRNA and form discrete nanoparticles. Importantly, it is demonstrated that the selection of the appropriate end-oligopeptide modifications enables the specific targeting and major transfection of antigen-presenting cells (APC) in vivo, after intravenous administration, thus enabling their use for immunotherapy strategies. Therefore, with this study, it can be confirmed that OM-PBAE are appropriate systems for the design of mRNA-based immunotherapy approaches aimed to in vivo transfect APCs and trigger immune responses to fight either tumors or infectious diseases.


Subject(s)
Antigen-Presenting Cells/metabolism , RNA, Messenger/administration & dosage , RNA, Messenger/metabolism , Animals , Cell Line , Cell Survival , Drug Carriers/chemistry , HeLa Cells , Humans , Immunotherapy , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Polymers/chemistry , RAW 264.7 Cells
4.
Mol Ther Nucleic Acids ; 8: 395-403, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28918039

ABSTRACT

The use of non-viral procedures, together with CRISPR/Cas9 genome-editing technology, allows the insertion of single-copy therapeutic genes at pre-determined genomic sites, overcoming safety limitations resulting from random gene insertions of viral vectors with potential for genome damage. In this study, we demonstrate that combination of non-viral gene delivery and CRISPR/Cas9-mediated knockin via homology-directed repair can replace the use of viral vectors for the generation of genetically modified therapeutic cells. We custom-modified human adipose mesenchymal stem cells (hAMSCs), using electroporation as a transfection method and CRISPR/Cas9-mediated knockin for the introduction and stable expression of a 3 kb DNA fragment including the eGFP (selectable marker) and a variant of the herpes simplex virus 1 thymidine kinase genes (therapeutic gene), under the control of the human elongation factor 1 alpha promoter in exon 5 of the endogenous thymidine kinase 2 gene. Using a U87 glioma model in SCID mice, we show that the therapeutic capacity of the new CRISPR/Cas9-engineered hAMSCs is equivalent to that of therapeutic hAMSCs generated by introduction of the same therapeutic gene by transduction with a lentiviral vector previously published by our group. This strategy should be of general use to other applications requiring genetic modification of therapeutic cells.

5.
Oncotarget ; 7(32): 51875-51897, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27391434

ABSTRACT

Metabolic reprogramming, a crucial cancer hallmark, shifts metabolic pathways such as glycolysis, tricarboxylic acid cycle or lipogenesis, to enable the growth characteristics of cancer cells. Here, we provide evidence that transketolase-like 1 (TKTL1) orchestrates aerobic glycolysis, fatty acid and nucleic acid synthesis, glutamine metabolism, protection against oxidative stress and cell proliferation. Furthermore, silencing of TKTL1 reduced the levels of sphingolipids such as lactosylceramide (a sphingolipid regulating cell survival, proliferation and angiogenesis) and phosphatidylinositol (which activates PI3K/Akt/mTOR signaling). Thus, in addition to its well-known roles in glucose and amino acid metabolism, TKTL1 also regulates lipid metabolism. In conclusion, our study provides unprecedented evidence that TKTL1 plays central roles in major metabolic processes subject to reprogramming in cancer cells and thus identifies TKTL1 as a promising target for new anti-cancer therapies.


Subject(s)
Metabolome , Neoplasms/metabolism , Transketolase/metabolism , Cell Line, Tumor , Glycolysis , Humans
6.
Tissue Eng Part C Methods ; 22(9): 864-72, 2016 09.
Article in English | MEDLINE | ID: mdl-27339005

ABSTRACT

Bioreactor systems allow safe and reproducible production of tissue constructs and functional analysis of cell behavior in biomaterials. However, current procedures for the analysis of tissue generated in biomaterials are destructive. We describe a transparent perfusion system that allows real-time bioluminescence imaging of luciferase expressing cells seeded in scaffolds for the study of cell-biomaterial interactions and bioreactor performance. A prototype provided with a poly(lactic) acid scaffold was used for "proof of principle" studies to monitor cell survival in the scaffold (up to 22 days). Moreover, using cells expressing a luciferase reporter under the control of inducible tissue-specific promoters, it was possible to monitor changes in gene expression resulting from hypoxic state and endothelial cell differentiation. This system should be useful in numerous tissue engineering applications, the optimization of bioreactor operation conditions, and the analysis of cell behavior in three-dimensional scaffolds.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Cell Proliferation , Image Processing, Computer-Assisted/methods , Luminescent Measurements , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Adipose Tissue/metabolism , Cell Culture Techniques , Humans , Mesenchymal Stem Cells/metabolism , Perfusion , Tissue Scaffolds
7.
Stem Cells ; 34(5): 1163-76, 2016 05.
Article in English | MEDLINE | ID: mdl-27146024

ABSTRACT

In solid tumors, cancer stem cells (CSCs) can arise independently of epithelial-mesenchymal transition (EMT). In spite of recent efforts, the metabolic reprogramming associated with CSC phenotypes uncoupled from EMT is poorly understood. Here, by using metabolomic and fluxomic approaches, we identify major metabolic profiles that differentiate metastatic prostate epithelial CSCs (e-CSCs) from non-CSCs expressing a stable EMT. We have found that the e-CSC program in our cellular model is characterized by a high plasticity in energy substrate metabolism, including an enhanced Warburg effect, a greater carbon and energy source flexibility driven by fatty acids and amino acid metabolism and an essential reliance on the proton buffering capacity conferred by glutamine metabolism. An analysis of transcriptomic data yielded a metabolic gene signature for our e-CSCs consistent with the metabolomics and fluxomics analyses that correlated with tumor progression and metastasis in prostate cancer and in 11 additional cancer types. Interestingly, an integrated metabolomics, fluxomics, and transcriptomics analysis allowed us to identify key metabolic players regulated at the post-transcriptional level, suggesting potential biomarkers and therapeutic targets to effectively forestall metastasis. Stem Cells 2016;34:1163-1176.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Metabolomics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Amino Acids/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Disease Progression , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Fatty Acids/biosynthesis , Gene Expression Profiling , Genes, Neoplasm , Glucose/metabolism , Glycolysis/drug effects , Glycolysis/genetics , Humans , Hydrogen-Ion Concentration , Mesoderm/pathology , Mitochondria/drug effects , Mitochondria/metabolism , NADP/metabolism , Neoplastic Stem Cells/drug effects , Oxidative Stress/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription, Genetic/drug effects
8.
Mol Cancer ; 13: 237, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25331979

ABSTRACT

BACKGROUND: Tumor cell subpopulations can either compete with each other for nutrients and physical space within the tumor niche, or co-operate for enhanced survival, or replicative or metastatic capacities. Recently, we have described co-operative interactions between two clonal subpopulations derived from the PC-3 prostate cancer cell line, in which the invasiveness of a cancer stem cell (CSC)-enriched subpopulation (PC-3M, or M) is enhanced by a non-CSC subpopulation (PC-3S, or S), resulting in their accelerated metastatic dissemination. METHODS: M and S secretomes were compared by SILAC (Stable Isotope Labeling by Aminoacids in Cell Culture). Invasive potential in vitro of M cells was analyzed by Transwell-Matrigel assays. M cells were co-injected with S cells in the dorsal prostate of immunodeficient mice and monitored by bioluminescence for tumor growth and metastatic dissemination. SPARC levels were determined by immunohistochemistry and real-time RT-PCR in tumors and by ELISA in plasma from patients with metastatic or non-metastatic prostate cancer. RESULTS: Comparative secretome analysis yielded 213 proteins differentially secreted between M and S cells. Of these, the protein most abundantly secreted in S relative to M cells was SPARC. Immunodepletion of SPARC inhibited the enhanced invasiveness of M induced by S conditioned medium. Knock down of SPARC in S cells abrogated the capacity of its conditioned medium to enhance the in vitro invasiveness of M cells and compromised their potential to boost the metastatic behavior of M cells in vivo. In most primary human prostate cancer samples, SPARC was expressed in the epithelial tumoral compartment of metastatic cases. CONCLUSIONS: The matricellular protein SPARC, secreted by a prostate cancer clonal tumor cell subpopulation displaying non-CSC properties, is a critical mediator of paracrine effects exerted on a distinct tumor cell subpopulation enriched in CSC. This paracrine interaction results in an enhanced metastatic behavior of the CSC-enriched tumor subpopulation. SPARC is expressed in the neoplastic cells of primary prostate cancer samples from metastatic cases, and could thus constitute a tumor progression biomarker and a therapeutic target in advanced prostate cancer.


Subject(s)
Lymphatic Metastasis/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Osteonectin/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Epithelium/drug effects , Epithelium/pathology , Extracellular Space/metabolism , Humans , Male , Neoplasm Invasiveness
9.
J Lipid Res ; 54(5): 1207-20, 2013 May.
Article in English | MEDLINE | ID: mdl-23423838

ABSTRACT

Acid ceramidase (AC) catalyzes the hydrolysis of ceramide into sphingosine, in turn a substrate of sphingosine kinases that catalyze its conversion into the mitogenic sphingosine-1-phosphate. AC is expressed at high levels in several tumor types and has been proposed as a cancer therapeutic target. Using a model derived from PC-3 prostate cancer cells, the highly tumorigenic, metastatic, and chemoresistant clone PC-3/Mc expressed higher levels of the AC ASAH1 than the nonmetastatic clone PC-3/S. Stable knockdown of ASAH1 in PC-3/Mc cells caused an accumulation of ceramides, inhibition of clonogenic potential, increased requirement for growth factors, and inhibition of tumorigenesis and lung metastases. We developed de novo ASAH1 inhibitors, which also caused a dose-dependent accumulation of ceramides in PC-3/Mc cells and inhibited their growth and clonogenicity. Finally, immunohistochemical analysis of primary prostate cancer samples showed that higher levels of ASAH1 were associated with more advanced stages of this neoplasia. These observations confirm ASAH1 as a therapeutic target in advanced and chemoresistant forms of prostate cancer and suggest that our new potent and specific AC inhibitors could act by counteracting critical growth properties of these highly aggressive tumor cells.


Subject(s)
Acid Ceramidase/antagonists & inhibitors , Acid Ceramidase/genetics , Molecular Targeted Therapy , Prostatic Neoplasms/genetics , Acid Ceramidase/metabolism , Apoptosis/genetics , Cell Line, Tumor , Ceramides/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lysophospholipids/metabolism , Male , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Sphingosine/analogs & derivatives , Sphingosine/metabolism
10.
J Clin Invest ; 122(5): 1849-68, 2012 May.
Article in English | MEDLINE | ID: mdl-22505459

ABSTRACT

Malignant progression in cancer requires populations of tumor-initiating cells (TICs) endowed with unlimited self renewal, survival under stress, and establishment of distant metastases. Additionally, the acquisition of invasive properties driven by epithelial-mesenchymal transition (EMT) is critical for the evolution of neoplastic cells into fully metastatic populations. Here, we characterize 2 human cellular models derived from prostate and bladder cancer cell lines to better understand the relationship between TIC and EMT programs in local invasiveness and distant metastasis. The model tumor subpopulations that expressed a strong epithelial gene program were enriched in highly metastatic TICs, while a second subpopulation with stable mesenchymal traits was impoverished in TICs. Constitutive overexpression of the transcription factor Snai1 in the epithelial/TIC-enriched populations engaged a mesenchymal gene program and suppressed their self renewal and metastatic phenotypes. Conversely, knockdown of EMT factors in the mesenchymal-like prostate cancer cell subpopulation caused a gain in epithelial features and properties of TICs. Both tumor cell subpopulations cooperated so that the nonmetastatic mesenchymal-like prostate cancer subpopulation enhanced the in vitro invasiveness of the metastatic epithelial subpopulation and, in vivo, promoted the escape of the latter from primary implantation sites and accelerated their metastatic colonization. Our models provide new insights into how dynamic interactions among epithelial, self-renewal, and mesenchymal gene programs determine the plasticity of epithelial TICs.


Subject(s)
Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Shape , Coculture Techniques , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Staging , Neoplasm Transplantation , Prostatic Neoplasms , Repressor Proteins/genetics , Repressor Proteins/metabolism , Snail Family Transcription Factors , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Urinary Bladder Neoplasms , Zinc Finger E-box-Binding Homeobox 1
11.
PLoS One ; 5(6): e11403, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20613989

ABSTRACT

BACKGROUND: Several pathways that control cell survival under stress, namely RNF8-dependent DNA damage recognition and repair, PCNA-dependent DNA damage tolerance and activation of NF-kappaB by extrinsic signals, are regulated by the tagging of key proteins with lysine 63-based polyubiquitylated chains, catalyzed by the conserved ubiquitin conjugating heterodimeric enzyme Ubc13-Uev. METHODOLOGY/PRINCIPAL FINDINGS: By applying a selection based on in vivo protein-protein interaction assays of compounds from a combinatorial chemical library followed by virtual screening, we have developed small molecules that efficiently antagonize the Ubc13-Uev1 protein-protein interaction, inhibiting the enzymatic activity of the heterodimer. In mammalian cells, they inhibit lysine 63-type polyubiquitylation of PCNA, inhibit activation of NF-kappaB by TNF-alpha and sensitize tumor cells to chemotherapeutic agents. One of these compounds significantly inhibited invasiveness, clonogenicity and tumor growth of prostate cancer cells. CONCLUSIONS/SIGNIFICANCE: This is the first development of pharmacological inhibitors of non-canonical polyubiquitylation that show that these compounds produce selective biological effects with potential therapeutic applications.


Subject(s)
Proteins/metabolism , Ubiquitination , Animals , Catalysis , HeLa Cells , Humans , Mice , Models, Animal , Models, Molecular , NF-kappa B/metabolism , Protein Binding
12.
Int J Cancer ; 125(11): 2565-75, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19530240

ABSTRACT

HER3 (ERBB3) is a catalytically inactive pseudokinase of the HER receptor tyrosine kinase family, frequently overexpressed in prostate and other cancers. Aberrant expression and mutations of 2 other members of the family, EGFR and HER2, are key carcinogenic events in several types of tumors, and both are well- validated therapeutic targets. In this study, we show that HER3 is required to maintain the motile and invasive phenotypes of prostate (DU-145) and breast (MCF-7) cancer cells in response to the HER3 ligand neuregulin-1 (NRG-1), epidermal growth factor (EGF) and fetal bovine serum. Although MCF-7 breast cancer cells appeared to require HER3 as part of an autocrine response induced by EGF and FBS, the response of DU-145 prostate cancer cells to these stimuli, while requiring HER3, did not appear to involve autocrine stimulation of the receptor. DU-145 cells required the expression of HER3 for efficient clonogenicity in vitro in standard growth medium and for tumorigenicity in immunodeficient mice. These observations suggest that prostate cancer cells derived from tumors that overexpress HER3 are dependent on its expression for the maintenance of major attributes of neoplastic aggressiveness, with or without cognate ligand stimulation.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , Neuregulin-1/metabolism , Prostatic Neoplasms/pathology , Receptor, ErbB-3/physiology , Animals , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement , Colony-Forming Units Assay , Disease Progression , Female , Gene Expression Profiling , Humans , Immunoprecipitation , Male , Mice , Mice, SCID , Neoplasm Invasiveness , Neuregulin-1/genetics , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Wound Healing , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...