Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Front Oncol ; 12: 869485, 2022.
Article in English | MEDLINE | ID: mdl-35837106

ABSTRACT

Metastasis is the primary cause of death in patients with colorectal cancer (CRC), urging the need for preclinical models that recapitulate the metastatic process at the individual patient level. We used an orthotopic patient-derived xenograft (PDX) obtained through the direct implantation of freshly dissociated CRC cells in the colon of immunocompromised mice to model the metastatic process. Ortho-PDX engraftment was associated to a specific set of molecular features of the parental tumor, such as epithelial-to-mesenchymal transition (EMT), TGF-ß pathway activation, increased expression of stemness-associated factors and higher numbers of circulating tumor cells (CTCs) clusters expressing the metastatic marker CD44v6. A parallel analysis of orthotopic/metastatic xenografts and organoids showed that tumor cells underwent mesenchymal-to-epithelial transition at the metastatic site and that metastasis-derived organoids had increased chemotherapy resistance. These observations support the usefulness of ortho-PDX as a preclinical model to study metastasis-related features and provide preliminary evidence that EMT/stemness properties of primary colorectal tumors may be crucial for orthotopic tumor engraftment.

2.
Methods Mol Biol ; 2429: 555-565, 2022.
Article in English | MEDLINE | ID: mdl-35507189

ABSTRACT

Cancer stem cells (CSCs) are responsible for the initiation of primary tumors and for metastasis seeding at distant organs. Therefore, they represent crucial targets for the study and preclinical testing of new antimetastatic approaches. We recently generated a molecularly characterized biobank of colorectal CSCs, isolated from individual patients and cultured in serum-free medium as multicellular spheroids. Here, we describe in detail the generation of a metastatic model of colorectal cancer based on the orthotopic injection of CSCs into the cecum serosa of immunodeficient mice. Such a model represents an excellent experimental system to investigate the cellular and molecular mechanisms involved in colorectal cancer metastasis, to analyze rare premetastatic elements such as circulating and disseminated tumor cells, and for the preclinical testing of new agents with potential antimetastatic activity.


Subject(s)
Colorectal Neoplasms , Neoplastic Stem Cells , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Heterografts , Humans , Mice , Neoplastic Stem Cells/pathology , Spheroids, Cellular/pathology
3.
J Exp Clin Cancer Res ; 41(1): 86, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35260172

ABSTRACT

BACKGROUND: Circulating tumor cells (CTCs) are responsible for the metastatic dissemination of colorectal cancer (CRC) to the liver, lungs and lymph nodes. CTCs rarity and heterogeneity strongly limit the elucidation of their biological features, as well as preclinical drug sensitivity studies aimed at metastasis prevention. METHODS: We generated organoids from CTCs isolated from an orthotopic CRC xenograft model. CTCs-derived organoids (CTCDOs) were characterized through proteome profiling, immunohistochemistry, immunofluorescence, flow cytometry, tumor-forming capacity and drug screening assays. The expression of intra- and extracellular markers found in CTCDOs was validated on CTCs isolated from the peripheral blood of CRC patients. RESULTS: CTCDOs exhibited a hybrid epithelial-mesenchymal transition (EMT) state and an increased expression of stemness-associated markers including the two homeobox transcription factors Goosecoid and Pancreatic Duodenal Homeobox Gene-1 (PDX1), which were also detected in CTCs from CRC patients. Functionally, CTCDOs showed a higher migratory/invasive ability and a different response to pathway-targeted drugs as compared to xenograft-derived organoids (XDOs). Specifically, CTCDOs were more sensitive than XDOs to drugs affecting the Survivin pathway, which decreased the levels of Survivin and X-Linked Inhibitor of Apoptosis Protein (XIAP) inducing CTCDOs death. CONCLUSIONS: These results indicate that CTCDOs recapitulate several features of colorectal CTCs and may be used to investigate the features of metastatic CRC cells, to identify new prognostic biomarkers and to devise new potential strategies for metastasis prevention.


Subject(s)
Colorectal Neoplasms , Neoplastic Cells, Circulating , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/physiology , Humans , Neoplastic Cells, Circulating/metabolism , Organoids/metabolism , Stem Cells/metabolism
4.
Cancers (Basel) ; 13(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34885020

ABSTRACT

Colorectal cancer (CRC) represents one of the most deadly cancers worldwide. Colorectal cancer stem cells (cCSCs) are the driving units of CRC initiation and development. After the concept of cCSC was first formulated in 2007, a huge bulk of research has contributed to expanding its definition, from a cell subpopulation defined by a fixed phenotype in a plastic entity modulated by complex interactions with the tumor microenvironment, in which cell position and niche-driven signals hold a prominent role. The wide development of cellular and molecular technologies recent years has been a main driver of advancements in cCSCs research. Here, we will give an overview of the parallel role of technological progress and of theoretical evolution in shaping the concept of cCSCs.

5.
Cell Death Differ ; 28(7): 2060-2082, 2021 07.
Article in English | MEDLINE | ID: mdl-33531658

ABSTRACT

Cancer stem cells (CSCs) are tumor subpopulations driving disease development, progression, relapse and therapy resistance, and their targeting ensures tumor eradication. CSCs display heterogeneous replication stress (RS), but the functionality/relevance of the RS response (RSR) centered on the ATR-CHK1 axis is debated. Here, we show that the RSR is efficient in primary CSCs from colorectal cancer (CRC-SCs), and describe unique roles for PARP1 and MRE11/RAD51. First, we demonstrated that PARP1 is upregulated in CRC-SCs resistant to several replication poisons and RSR inhibitors (RSRi). In these cells, PARP1 modulates replication fork speed resulting in low constitutive RS. Second, we showed that MRE11 and RAD51 cooperate in the genoprotection and mitosis execution of PARP1-upregulated CRC-SCs. These roles represent therapeutic vulnerabilities for CSCs. Indeed, PARP1i sensitized CRC-SCs to ATRi/CHK1i, inducing replication catastrophe, and prevented the development of resistance to CHK1i. Also, MRE11i + RAD51i selectively killed PARP1-upregulated CRC-SCs via mitotic catastrophe. These results provide the rationale for biomarker-driven clinical trials in CRC using distinct RSRi combinations.


Subject(s)
Colorectal Neoplasms/drug therapy , MRE11 Homologue Protein/drug effects , Mitosis/drug effects , Neoplastic Stem Cells/drug effects , Poly (ADP-Ribose) Polymerase-1/drug effects , Rad51 Recombinase/drug effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA Replication/drug effects , Humans , MRE11 Homologue Protein/genetics , Neoplastic Stem Cells/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Rad51 Recombinase/genetics
6.
Front Oncol ; 10: 592891, 2020.
Article in English | MEDLINE | ID: mdl-33194755

ABSTRACT

Severe coronavirus disease 2019 (COVID-19) causes an uncontrolled activation of the innate immune response, resulting in acute respiratory distress syndrome and systemic inflammation. The effects of COVID-19-induced inflammation on cancer cells and their microenvironment are yet to be elucidated. Here, we formulate the hypothesis that COVID-19-associated inflammation may generate a microenvironment favorable to tumor cell proliferation and particularly to the reawakening of dormant cancer cells (DCCs). DCCs often survive treatment of primary tumors and populate premetastatic niches in the lungs and other organs, retaining the potential for metastatic outgrowth. DCCs reawakening may be promoted by several events associated to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, including activation of neutrophils and monocytes/macrophages, lymphopenia and an uncontrolled production of pro-inflammatory cytokines. Among pro-inflammatory factors produced during COVID-19, neutrophil extracellular traps (NETs) released by activated neutrophils have been specifically shown to activate premetastatic cancer cells disseminated in the lungs, suggesting they may be involved in DCCs reawakening in COVID-19 patients. If confirmed by further studies, the links between COVID-19, DCCs reactivation and tumor relapse may support the use of specific anti-inflammatory and anti-metastatic therapies in patients with COVID-19 and an active or previous cancer.

7.
J Exp Clin Cancer Res ; 39(1): 2, 2020 Jan 08.
Article in English | MEDLINE | ID: mdl-31910865

ABSTRACT

BACKGROUND: Quiescent/slow cycling cells have been identified in several tumors and correlated with therapy resistance. However, the features of chemoresistant populations and the molecular factors linking quiescence to chemoresistance are largely unknown. METHODS: A population of chemoresistant quiescent/slow cycling cells was isolated through PKH26 staining (which allows to separate cells on the basis of their proliferation rate) from colorectal cancer (CRC) xenografts and subjected to global gene expression and pathway activation analyses. Factors expressed by the quiescent/slow cycling population were analyzed through lentiviral overexpression approaches for their ability to induce a dormant chemoresistant state both in vitro and in mouse xenografts. The correlation between quiescence-associated factors, CRC consensus molecular subtype and cancer prognosis was analyzed in large patient datasets. RESULTS: Untreated colorectal tumors contain a population of quiescent/slow cycling cells with stem cell features (quiescent cancer stem cells, QCSCs) characterized by a predetermined mesenchymal-like chemoresistant phenotype. QCSCs expressed increased levels of ZEB2, a transcription factor involved in stem cell plasticity and epithelial-mesenchymal transition (EMT), and of antiapototic factors pCRAF and pASK1. ZEB2 overexpression upregulated pCRAF/pASK1 levels resulting in increased chemoresistance, enrichment of cells with stemness/EMT traits and proliferative slowdown of tumor xenografts. In parallel, chemotherapy treatment of tumor xenografts induced the prevalence of QCSCs with a stemness/EMT phenotype and activation of the ZEB2/pCRAF/pASK1 axis, resulting in a chemotherapy-unresponsive state. In CRC patients, increased ZEB2 levels correlated with worse relapse-free survival and were strongly associated to the consensus molecular subtype 4 (CMS4) characterized by dismal prognosis, decreased proliferative rates and upregulation of EMT genes. CONCLUSIONS: These results show that chemotherapy-naive tumors contain a cell population characterized by a coordinated program of chemoresistance, quiescence, stemness and EMT. Such population becomes prevalent upon drug treatment and is responsible for chemotherapy resistance, thus representing a key target for more effective therapeutic approaches.


Subject(s)
Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Neoplastic Stem Cells/metabolism , Up-Regulation , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Female , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Kinase Kinase 5/metabolism , Male , Mice , Middle Aged , Neoplasm Transplantation , Oxaliplatin/pharmacology , Prognosis
8.
J Exp Clin Cancer Res ; 38(1): 373, 2019 Aug 22.
Article in English | MEDLINE | ID: mdl-31439019

ABSTRACT

BACKGROUND: An increasing number of anticancer agents has been proposed in recent years with the attempt to overcome treatment-resistant cancer cells and particularly cancer stem cells (CSC), the major culprits for tumour resistance and recurrence. However, a huge obstacle to treatment success is the ineffective delivery of drugs within the tumour environment due to limited solubility, short circulation time or inconsistent stability of compounds that, together with concomitant dose-limiting systemic toxicity, contribute to hamper the achievement of therapeutic drug concentrations. The synthetic retinoid Fenretinide (4-hydroxy (phenyl)retinamide; 4-HPR) formerly emerged as a promising anticancer agent based on pre-clinical and clinical studies. However, a major limitation of fenretinide is traditionally represented by its poor aqueous solubility/bioavailability due to its hydrophobic nature, that undermined the clinical success of previous clinical trials. METHODS: Here, we developed a novel nano-micellar fenretinide formulation called bionanofenretinide (Bio-nFeR), based on drug encapsulation in an ion-pair stabilized lipid matrix, with the aim to raise fenretinide bioavailability and antitumour efficacy. RESULTS: Bio-nFeR displayed marked antitumour activity against lung, colon and melanoma CSC both in vitro and in tumour xenografts, in absence of mice toxicity. Bio-nFeR is suitable for oral administration, reaching therapeutic concentrations within tumours and an unprecedented therapeutic activity in vivo as single agent. CONCLUSION: Altogether, our results indicate Bio-nFeR as a novel anticancer agent with low toxicity and high activity against tumourigenic cells, potentially useful for the treatment of solid tumours of multiple origin.


Subject(s)
Colonic Neoplasms/drug therapy , Fenretinide/administration & dosage , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Micelles , Neoplastic Stem Cells/drug effects , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis , Biological Availability , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Fenretinide/chemistry , Fenretinide/pharmacokinetics , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cell Death Dis ; 10(7): 529, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31332161

ABSTRACT

Fenretinide is a synthetic retinoid characterized by anticancer activity in preclinical models and favorable toxicological profile, but also by a low bioavailability that hindered its clinical efficacy in former clinical trials. We developed a new formulation of fenretinide complexed with 2-hydroxypropyl-beta-cyclodextrin (nanofenretinide) characterized by an increased bioavailability and therapeutic efficacy. Nanofenretinide was active in cell lines derived from multiple solid tumors, in primary spheroid cultures and in xenografts of lung and colorectal cancer, where it inhibited tumor growth independently from the mutational status of tumor cells. A global profiling of pathways activated by nanofenretinide was performed by reverse-phase proteomic arrays and lipid analysis, revealing widespread repression of the mTOR pathway, activation of apoptotic, autophagic and DNA damage signals and massive production of dihydroceramide, a bioactive lipid with pleiotropic effects on several biological processes. In cells that survived nanofenretinide treatment there was a decrease of factors involved in cell cycle progression and an increase in the levels of p16 and phosphorylated p38 MAPK with consequent block in G0 and early G1. The capacity of nanofenretinide to induce cancer cell death and quiescence, together with its elevated bioavailability and broad antitumor activity indicate its potential use in cancer treatment and chemoprevention.


Subject(s)
Antineoplastic Agents/therapeutic use , Fenretinide/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , DNA Damage/drug effects , DNA Damage/genetics , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
ACS Med Chem Lett ; 10(4): 499-503, 2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30996786

ABSTRACT

Targeted approaches aiming at modulating NHERF1 activity, rather than its overall expression, would be preferred to preserve the normal functions of this versatile protein. We focused our attention on the NHERF1/PDZ1 domain that governs its membrane recruitment/displacement through a transient phosphorylation switch. We herein report the design and synthesis of novel NHERF1 PDZ1 domain inhibitors. These compounds have potential therapeutic value when used in combination with antagonists of ß-catenin to augment apoptotic death of colorectal cancer cells refractory to currently available Wnt/ß-catenin-targeted agents.

11.
Cell Biol Toxicol ; 34(6): 459-469, 2018 12.
Article in English | MEDLINE | ID: mdl-29478126

ABSTRACT

Biobanking of molecularly characterized colorectal cancer stem cells (CSCs) generated from individual patients and growing as spheroids in defined serum-free media offer a fast, feasible, and multi-level approach for the screening of targeted therapies and drug resistance molecular studies. By combining in vitro and in vivo analyses of cetuximab efficacy with genetic data on an ongoing collection of stem cell-enriched spheroids, we describe the identification and preliminary characterization of microsatellite stable (MSS) CSCs that, despite the presence of the KRAS (G12D) mutation, display epidermal growth factor (EGF)-dependent growth and are strongly inhibited by anti-EGF-receptor (EGFR) treatment. In parallel, we detected an increased resistance to anti-EGFR therapy of microsatellite instable (MSI) CSC lines irrespective of KRAS mutational status. MSI CSC lines carried mutations in genes coding for proteins with a role in RAS and calcium signaling, highlighting the role of a genomically unstable context in determining anti-EGFR resistance. Altogether, these results argue for a multifactorial origin of anti-EGFR resistance that emerges as the effect of multiple events targeting direct and indirect regulators of the EGFR pathway. An improved understanding of key molecular determinants of sensitivity/resistance to EGFR inhibition will be instrumental to optimize the clinical efficacy of anti-EGFR agents, representing a further step towards personalized treatments.


Subject(s)
Colorectal Neoplasms/metabolism , Drug Evaluation, Preclinical/methods , Neoplastic Stem Cells/drug effects , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Biological Specimen Banks/trends , Cetuximab/pharmacology , Colorectal Neoplasms/physiopathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Epidermal Growth Factor/physiology , ErbB Receptors/physiology , Humans , Mutation , Panitumumab , Precision Medicine/methods , Proto-Oncogene Proteins p21(ras)/genetics , Spheroids, Cellular/physiology , Tumor Cells, Cultured/physiology
12.
Methods Mol Biol ; 1692: 107-115, 2018.
Article in English | MEDLINE | ID: mdl-28986891

ABSTRACT

Banks of genetically characterized cancer stem cells (CSCs) isolated from individual patients and grown as spheroids offer an invaluable approach to identify genetic determinants of drug resistance versus sensitivity, and to study new stem cell-directed therapies. Here, we describe our standardized procedure for in vitro drug screening on colorectal CSCs, taking irinotecan as an example.


Subject(s)
Colorectal Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Animals , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/physiology , Humans , Irinotecan
13.
Gut ; 67(5): 903-917, 2018 05.
Article in English | MEDLINE | ID: mdl-28389531

ABSTRACT

OBJECTIVE: Cancer stem cells (CSCs) are responsible for tumour formation and spreading, and their targeting is required for tumour eradication. There are limited therapeutic options for advanced colorectal cancer (CRC), particularly for tumours carrying RAS-activating mutations. The aim of this study was to identify novel CSC-targeting strategies. DESIGN: To discover potential therapeutics to be clinically investigated as single agent, we performed a screening with a panel of FDA-approved or investigational drugs on primary CRC cells enriched for CSCs (CRC-SCs) isolated from 27 patients. Candidate predictive biomarkers of efficacy were identified by integrating genomic, reverse-phase protein microarray (RPPA) and cytogenetic analyses, and validated by immunostainings. DNA replication stress (RS) was increased by employing DNA replication-perturbing or polyploidising agents. RESULTS: The drug-library screening led to the identification of LY2606368 as a potent anti-CSC agent acting in vitro and in vivo in tumour cells from a considerable number of patients (∼36%). By inhibiting checkpoint kinase (CHK)1, LY2606368 affected DNA replication in most CRC-SCs, including RAS-mutated ones, forcing them into premature, lethal mitoses. Parallel genomic, RPPA and cytogenetic analyses indicated that CRC-SCs sensitive to LY2606368 displayed signs of ongoing RS response, including the phosphorylation of RPA32 and ataxia telangiectasia mutated serine/threonine kinase (ATM). This was associated with mutation(s) in TP53 and hyperdiploidy, and made these CRC-SCs exquisitely dependent on CHK1 function. Accordingly, experimental increase of RS sensitised resistant CRC-SCs to LY2606368. CONCLUSIONS: LY2606368 selectively eliminates replication-stressed, p53-deficient and hyperdiploid CRC-SCs independently of RAS mutational status. These results provide a strong rationale for biomarker-driven clinical trials with LY2606368 in patients with CRC.


Subject(s)
Antineoplastic Agents/pharmacology , Checkpoint Kinase 1/drug effects , Colorectal Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Pyrazines/pharmacology , Pyrazoles/pharmacology , Cell Line, Tumor , Checkpoint Kinase 1/genetics , Colorectal Neoplasms/genetics , DNA Replication/drug effects , Humans , Immunohistochemistry , Mutation , Neoplastic Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Tumor Suppressor Protein p53/genetics
14.
EBioMedicine ; 20: 39-49, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28551381

ABSTRACT

8-Oxoguanine, a common mutagenic DNA lesion, generates G:C>T:A transversions via mispairing with adenine during DNA replication. When operating normally, the MUTYH DNA glycosylase prevents 8-oxoguanine-related mutagenesis by excising the incorporated adenine. Biallelic MUTYH mutations impair this enzymatic function and are associated with colorectal cancer (CRC) in MUTYH-Associated Polyposis (MAP) syndrome. Here, we perform whole-exome sequencing that reveals a modest mutator phenotype in MAP CRCs compared to sporadic CRC stem cell lines or bulk tumours. The excess G:C>T:A transversion mutations in MAP CRCs exhibits a novel mutational signature, termed Signature 36, with a strong sequence dependence. The MUTYH mutational signature reflecting persistent 8-oxoG:A mismatches occurs frequently in the APC, KRAS, PIK3CA, FAT4, TP53, FAT1, AMER1, KDM6A, SMAD4 and SMAD2 genes that are associated with CRC. The occurrence of Signature 36 in other types of human cancer indicates that DNA 8-oxoguanine-related mutations might contribute to the development of cancer in other organs.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Damage , DNA Glycosylases/genetics , Guanine/analogs & derivatives , Alleles , Colorectal Neoplasms/pathology , DNA Glycosylases/metabolism , DNA Mutational Analysis , DNA Repair , Gene Frequency , Genes, Tumor Suppressor , Genetic Association Studies , Genetic Predisposition to Disease , Guanine/metabolism , Humans , Microsatellite Instability , Mutation , Mutation Rate , Oncogenes , Exome Sequencing
15.
Stem Cells Transl Med ; 5(4): 511-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26956206

ABSTRACT

UNLABELLED: Colorectal cancer (CRC) therapy mainly relies on the use of conventional chemotherapeutic drugs combined, in a subset of patients, with epidermal growth factor receptor [EGFR]-targeting agents. Although CRC is considered a prototype of a cancer stem cell (CSC)-driven tumor, the effects of both conventional and targeted therapies on the CSC compartment are largely unknown. We have optimized a protocol for colorectal CSC isolation that allowed us to obtain CSC-enriched cultures from primary tumor specimens, with high efficiency. CSC isolation was followed by in vitro and in vivo validation, genetic characterization, and drug sensitivity analysis, thus generating panels of CSC lines with defined patterns of genetic mutations and therapy sensitivity. Colorectal CSC lines were polyclonal and maintained intratumor heterogeneity in terms of somatically acquired mutations and differentiation state. Such CSC-enriched cultures were used to investigate the effects of both conventional and targeted therapies on the CSC compartment in vivo and to generate a proteomic picture of signaling pathways implicated in sensitivity/resistance to anti-EGFR agents. We propose CSC lines as a sound preclinical framework to test the effects of therapies in vitro and in vivo and to identify novel determinants of therapy resistance. SIGNIFICANCE: Colorectal cancer stem cells (CSCs) have been shown to be responsible for tumor propagation, metastatic dissemination, and relapse. However, molecular pathways present in CSCs, as well as mechanisms of therapy resistance, are mostly unknown. Taking advantage of genetically characterized CSC lines derived from colorectal tumors, this study provides an extensive analysis of CSC response to EGFR-targeted therapy in vivo and an overview of factors implicated in therapy response or resistance. Furthermore, the implementation of a biobank of molecularly annotated CSC lines provides an innovative resource for future investigations in colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Neoplastic Stem Cells/pathology , Animals , Cells, Cultured , Female , Gene Expression Profiling , Humans , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Microarray Analysis , Models, Biological , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Proteomics , Signal Transduction/genetics
16.
PLoS One ; 9(4): e94438, 2014.
Article in English | MEDLINE | ID: mdl-24740347

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been reported to exhibit therapeutic activity in cancer. However, many tumors remain resistant to treatment with TRAIL. Therefore, small molecules that potentiate the cytotoxic effects of TRAIL could be used for combinatorial therapy. Here we found that the ionophore antibiotic salinomycin acts in synergism with TRAIL, enhancing TRAIL-induced apoptosis in glioma cells. Treatment with low doses of salinomycin in combination with TRAIL augmented the activation of caspase-3 and increased TRAIL-R2 cell surface expression. TRAIL-R2 upmodulation was required for mediating the stimulatory effect of salinomycin on TRAIL-mediated apoptosis, since it was abrogated by siRNA-mediated TRAIL-R2 knockdown. Salinomycin in synergism with TRAIL exerts a marked anti-tumor effect in nude mice xenografted with human glioblastoma cells. Our results suggest that the combination of TRAIL and salinomycin may be a useful tool to overcome TRAIL resistance in glioma cells and may represent a potential drug for treatment of these tumors. Importantly, salinomycin+TRAIL were able to induce cell death of well-defined glioblastoma stem-like lines.


Subject(s)
Cytotoxins/pharmacology , Glioblastoma/drug therapy , Pyrans/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Humans , Mice, Nude
17.
Stem Cells ; 30(9): 1819-30, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22753241

ABSTRACT

Tumor-initiating cells are responsible for tumor maintenance and relapse in solid and hematologic cancers. Although tumor-initiating cells were initially believed to be mainly quiescent, rapidly proliferating tumorigenic cells were found in breast cancer. In colon cancer, the proliferative activity of the tumorigenic population has not been defined, although it represents an essential parameter for the development of more effective therapeutic strategies. Here, we show that tumorigenic colon cancer cells can be found in a rapidly proliferating state in vitro and in vivo, both in human tumors and mouse xenografts. Inhibitors of polo-like kinase1 (Plk1), a mitotic kinase essential for cell proliferation, demonstrated maximal efficiency over other targeted compounds and chemotherapeutic agents in inducing death of colon cancer-initiating cells in vitro. In vivo, Plk1 inhibitors killed CD133(+) colon cancer cells leading to complete growth arrest of colon cancer stem cell-derived xenografts, whereas chemotherapeutic agents only slowed tumor progression. While chemotherapy treatment increased CD133(+) cell proliferation, treatment with Plk1 inhibitors eliminated all proliferating tumor-initiating cells. Quiescent CD133(+) cells that survived the treatment with Plk1 inhibitors could be killed by subsequent Plk1 inhibition when they exited from quiescence. Altogether, these results provide a new insight into the proliferative status of colon tumor-initiating cells both in basal conditions and in response to therapy and indicate Plk1 inhibitors as potentially useful in the treatment of colorectal cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , AC133 Antigen , Animals , Antigens, CD/biosynthesis , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Colonic Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic , Glycoproteins/biosynthesis , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mitochondria/drug effects , Mitochondria/physiology , Peptides , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Pteridines/pharmacology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transplantation, Heterologous , Polo-Like Kinase 1
18.
Mol Reprod Dev ; 79(2): 118-27, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22139884

ABSTRACT

Two large families of retrotransposons, that is, LINE-1 (Long Interspersed Nuclear Elements-1) and endogenous retroviruses, encode reverse transcriptase (RT) proteins in vertebrates. We previously showed that mouse preimplantation embryos are endowed with an endogenous, functional RT activity. Inhibiting that activity by microinjecting antisense oligonucleotides against a highly active LINE-1 family member in mouse oocytes blocked developmental progression between the two- and four-blastomere stages, indicating that LINE-1-encoded RT activity is strictly required at this critical transition in early development. Here we show that incubation of mouse zygotes with 5'-bromodeoxyuridine (BrdU) yields massive incorporation of this nucleoside analogue in newly synthesized DNA; surprisingly, a significant incorporation still occurs in both zygotic pronuclei in the presence of aphidicolin, a specific inhibitor of DNA replication. This aphidicolin-resistant BrdU incorporation is quantitatively abolished when embryos are simultaneously exposed to abacavir, a nucleoside RT inhibitor, indicating its retrotranscription-dependent nature. Moreover, quantitative PCR analysis revealed a burst of new synthesis of LINE-1 copies at the zygote- and two-cell embryo stages. These findings support the conclusion that RT-dependent amplification of LINE-1 retrotransposons is a distinctive feature of early embryonic genomes. Its physiological involvement in preimplantation murine development is discussed.


Subject(s)
DNA Copy Number Variations , Embryonic Development/genetics , Gene Amplification , Long Interspersed Nucleotide Elements/genetics , Animals , Bromodeoxyuridine/pharmacokinetics , Cell Nucleus/metabolism , Cleavage Stage, Ovum/metabolism , Cleavage Stage, Ovum/physiology , DNA Copy Number Variations/physiology , DNA Replication/genetics , Embryo, Mammalian , Female , Gene Amplification/physiology , Gene Expression Regulation, Developmental , Male , Mice , Retroelements/genetics , Zygote/metabolism
19.
Curr Opin Pharmacol ; 10(4): 380-4, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20561817

ABSTRACT

Human cancer stem cells represent promising tools for new approaches to pathway-targeted drug discovery and preclinical screening. Because of their distinctive capability to recapitulate the development of the original tumors in vivo, the study of human cancer stem cells (CSCs) largely relies on models of xenograft transplantation into immunodeficient mice. In this frame, immunity and microenvironment-related issues need careful consideration. Here we shortly revise present knowledge on xenograft-related aspects of human CSCs studies.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms, Experimental/drug therapy , Neoplastic Stem Cells/drug effects , Animals , Cell Line, Tumor , Drug Discovery , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Xenograft Model Antitumor Assays
20.
Exp Hematol ; 36(8): 1035-46, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18504067

ABSTRACT

Mesenchymal stromal cells (MSCs) represent a bone marrow (BM) population, classically defined by five functional properties: extensive proliferation, ability to differentiate into osteoblasts, chondrocytes, adipocytes, and stromal cells-supporting hematopoiesis. However, research progress in this area has been hampered by lack of suitable markers and standardized procedures for MSC isolation. We have isolated a CD146(+) multipotent MSC population from 20 human BM donors displaying the phenotype of self-renewing osteoprogenitors; an extensive 12-week proliferation; and the ability to differentiate in osteoblasts, chondrocytes, adipocytes, and stromal cells supporting hematopoiesis. Furthermore, the CD146(+) MSCs secrete a complex combination of growth factors (GFs) controlling hematopoietic stem cells (HSCs) function, while providing a >2-log increase in the long-term culture (LTC) colony output in 8-week LTC over conventional assays. The hematopoietic stromal function exhibited by the MSCs was further characterized by manipulating LTCs with the chemical inhibitors Imatinib or SU-5416, targeting two GF receptors (GFRs), KIT or VEGFR2/1, respectively. Both treatments similarly impaired LTC colony output, indicating key roles for these two GF/GFR interactions to support LTC-initiating cell activity. CD146(+) MSCs may thus represent a tool to explore the MSC-HSC cross-talk in an in vitro surrogate model for HSC "niches," and for regenerative therapy studies. In addition, the MSC microRNA (miRNA) expression profile was analyzed by microarrays in both basic conditions and chondrogenic differentiation. Our analysis revealed that several miRNAs are modulated during chondrogenesis, and many of their putative targets are genes involved in chondrogenic differentiation.


Subject(s)
CD146 Antigen/biosynthesis , Cell Line , Mesenchymal Stem Cells/cytology , MicroRNAs/biosynthesis , Stromal Cells/cytology , Blotting, Western , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cell Separation/methods , Chondrocytes/cytology , Gene Expression Profiling , Humans , Immunophenotyping , Intercellular Signaling Peptides and Proteins/biosynthesis , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Microarray Analysis , Molecular Sequence Data , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...