43 results on '"E. Lonardo"'
Search Results
2. Metropolis. Urban Mindscapes
- Author
-
E. Lonardo
- Subjects
Psychogeography ,Mindscapes, Urban psychology, Urban interiors, Urban design, Placemaking, Psychogeography ,Placemaking ,Point (typography) ,media_common.quotation_subject ,Urban design ,Urban psychology ,Psyche ,State (polity) ,Aesthetics ,Sociology ,Mindscapes ,Urban interiors ,Urban space ,media_common - Abstract
In the present chapter, a contribution will try to be offered to the debate on the project of contemporary cities, through a trans-disciplinary approach, starting from a central point of view that often, when dealing with a project of urban space, is taken into marginal account: the psychological one. Is the psyche of contemporary human beings sufficiently prepared to live well in the cities we have built? Does it make sense to talk about smart cities, if people are still expecting to see their basic individual and collective needs resolved in the urban fabric? The text starts from an examination of the research state of the art, then continues with an analysis of the characteristics that contribute to recognizing an urban space as a place, highlighting a series of behaviors that could be deepened or used as a driver for the re-appropriation of the city.
- Published
- 2020
3. Peer Review #3 of 'Co-expression of HIF-1α, MDR1 and LAPTM4B in peripheral blood of solid tumors (v0.2)'
- Author
-
E Lonardo
- Subjects
LAPTM4B ,business.industry ,Cancer research ,Medicine ,business ,Peripheral blood - Published
- 2019
- Full Text
- View/download PDF
4. List of Contributors
- Author
-
A. Ahmad, R. Ankrah, M.A. Aziz, M.H. Aziz, S.W. Aziz, G. Bassi, M. Burke, A.F. Chambers, S. Chandradas, P. Chanvorachote, J.L. Chin, D. Chitale, P. Chunhacha, K. Debiec, M. El-Tanani, S. El-Tanani, P.J. Foster, C.T. Frenette, M. Garancini, I. Garcia-Diez, M. Gonzalez, S. Iwata, M. Krampera, T. Krueger, D.-Q. Li, P.M. Loadman, E. Lonardo, P. Martinelli, S.F. Miler, R. Morgan, J.M. Muller, D.H. Murrell, S.D. Nathanson, C. Nicholson, A.H. Nwabo Kamdje, L. Pattterson, J.Y. Perentes, F. Perera, E. Pinotti, J.A. Plock, F. Romano, K. Rosso, P.S. Rudland, P.F. Seke Etet, Z.-M. Shao, Y. Shiozawa, K.M. Siddiqui, C.B. Skillin, P. Takam Kamga, C.Y. Thomas, A. Toll, K.-C. Tran, W. Tsuji, F. Uggeri, L. Vecchio, I. Vela, E.D. Williams, J. Wydmanski, and H. Zubair
- Published
- 2017
- Full Text
- View/download PDF
5. Unraveling the role of EGF-CFC Cripto in muscle regeneration
- Author
-
O. GUARDIOLA, P. LAFUSTE, E. Lonardo, P. CHEVRON, A. CARTON, L. MOONS, P. CARMELIET, and G.MINCHIOTTI.
- Published
- 2009
6. Method for Differentiation of Multiple Neuronal Subtypes from Mouse Embryonic Stem Cells
- Author
-
S. Parisi, E. Lonardo, A. Fico, S. Filosa, and G. Minchiotti
- Published
- 2008
7. IDENTIFICATION AND CHARACTERIZATION OF MOLECULES INVOLVED IN NEURAL DIFFERENTIATION OF STEM CELLS: IMPROVING THE USE OF CELL-BASED THERAPY IN NEURODEGENERATIVE DISORDERS
- Author
-
E. Lonardo, C. Parish, S. Ponticelli, D. Marasco, S. De Falco, M. Ruvo, E. Arenas, and G. Minchiotti
- Published
- 2007
8. [Myocardial T1-201 scintigraphy in the study of the development of acute myocardial infarction. Evaluation of the effects of a drug with metabolic action: creatine phosphate]
- Author
-
V, Pedone, C, Corbelli, C, Frondini, R, Franchi, M, Luppi, E, Lonardo, A, Balboni, N, Monetti, and G, di Biase
- Subjects
Adult ,Radioisotopes ,Phosphocreatine ,Infant, Newborn ,Myocardial Infarction ,Humans ,Middle Aged ,Thallium ,Radionuclide Imaging ,Aged - Published
- 1984
9. Hybrid Biosilica Nanoparticles for in-vivo Targeted Inhibition of Colorectal Cancer Growth and Label-Free Imaging.
- Author
-
Delle Cave D, Mangini M, Tramontano C, De Stefano L, Corona M, Rea I, De Luca AC, and Lonardo E
- Subjects
- Animals, Humans, Mice, Silicon Dioxide chemistry, Cell Line, Tumor, Epithelial-Mesenchymal Transition drug effects, Neural Cell Adhesion Molecule L1 antagonists & inhibitors, Neural Cell Adhesion Molecule L1 chemistry, Transforming Growth Factor beta antagonists & inhibitors, Pyrazoles chemistry, Pyrazoles pharmacology, Pyrazoles administration & dosage, Spectrum Analysis, Raman, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, Colorectal Neoplasms diagnostic imaging, Gold chemistry, Gold pharmacology, Metal Nanoparticles chemistry
- Abstract
Background: Metastasis-initiating cells are key players in progression, resistance, and relapse of colorectal cancer (CRC), by leveraging the regulatory relationship between Transforming Growth Factor-beta (TGF-β) signaling and anti-L1 cell adhesion molecule (L1CAM)., Methods: This study introduces a novel strategy for CRC targeted therapy and imaging based on the use of a hybrid nanosystem made of gold nanoparticles-covered porous biosilica further modified with the (L1CAM) antibody., Results: The nanosystem intracellularly delivers galunisertib (LY), a TGF-β inhibitor, aiming to inhibit epithelial-mesenchymal transition (EMT), a process pivotal for metastasis. Anti-L1CAM antibody-functionalized nanoparticles (NPs) target tumor-initiating cells expressing L1CAM, inhibiting cancer growth. The number of antibody molecules conjugated to the single NP is precisely quantified, revealing a high surface coverage that facilitates the tumor targeting. The therapeutic efficacy of the nanosystem is investigated in organoid-like cultures of CRC cells and in vivo mouse models, showing a significant reduction in tumor growth. The spatial distribution of NPs within CRC tumors from mice is investigated using a label-free optical approach based on Raman micro-spectroscopy., Conclusion: This research highlights the multifunctional capabilities of engineered biosilica NPs, which offer new insights in targeted CRC therapy and imaging, improving patient outcomes and paving the way for personalized therapies., Competing Interests: Chiara Tramontano is currently affiliated with Internal Medicine Department, Radboud University Medical Center, Radboud, The Netherlands. The authors report no conflict of interest in this work., (© 2024 Delle Cave et al.)
- Published
- 2024
- Full Text
- View/download PDF
10. A 3D Pancreatic Cancer Model with Integrated Optical Sensors for Noninvasive Metabolism Monitoring and Drug Screening.
- Author
-
Siciliano AC, Forciniti S, Onesto V, Iuele H, Cave DD, Carnevali F, Gigli G, Lonardo E, and Del Mercato LL
- Subjects
- Humans, Apoptosis drug effects, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Hydrogen-Ion Concentration, Microgels, Stromal Cells drug effects, Tumor Microenvironment drug effects, Gemcitabine pharmacology, Paclitaxel pharmacology, Antineoplastic Combined Chemotherapy Protocols, Drug Screening Assays, Antitumor methods, Pancreatic Neoplasms chemistry, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Antineoplastic Agents pharmacology, Models, Biological
- Abstract
A distinct feature of pancreatic ductal adenocarcinoma (PDAC) is a prominent tumor microenvironment (TME) with remarkable cellular and spatial heterogeneity that meaningfully impacts disease biology and treatment resistance. The dynamic crosstalk between cancer cells and the dense stromal compartment leads to spatially and temporally heterogeneous metabolic alterations, such as acidic pH that contributes to drug resistance in PDAC. Thus, monitoring the extracellular pH metabolic fluctuations within the TME is crucial to predict and to quantify anticancer drug efficacy. Here, a simple and reliable alginate-based 3D PDAC model embedding ratiometric optical pH sensors and cocultures of tumor (AsPC-1) and stromal cells for simultaneously monitoring metabolic pH variations and quantify drug response is presented. By means of time-lapse confocal laser scanning microscopy (CLSM) coupled with a fully automated computational analysis, the extracellular pH metabolic variations are monitored and quantified over time during drug testing with gemcitabine, folfirinox, and paclitaxel, commonly used in PDAC therapy. In particular, the extracellular acidification is more pronounced after drugs treatment, resulting in increased antitumor effect correlated with apoptotic cell death. These findings highlight the importance of studying the influence of cellular metabolic mechanisms on tumor response to therapy in 3D tumor models, this being crucial for the development of personalized medicine approaches., (© 2024 Wiley‐VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
11. Three-dimensional environment sensitizes pancreatic cancer cells to the anti-proliferative effect of budesonide by reprogramming energy metabolism.
- Author
-
Ibello E, Saracino F, Delle Cave D, Buonaiuto S, Amoroso F, Andolfi G, Corona M, Guardiola O, Colonna V, Sainz B Jr, Altucci L, De Cesare D, Cobellis G, Lonardo E, Patriarca EJ, D'Aniello C, and Minchiotti G
- Subjects
- Humans, Mice, Animals, Cell Line, Tumor, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Xenograft Model Antitumor Assays, Cell Movement drug effects, Budesonide pharmacology, Budesonide therapeutic use, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Energy Metabolism drug effects, Cell Proliferation drug effects
- Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is the most lethal cancer with an aggressive metastatic phenotype and very poor clinical prognosis. Interestingly, a lower occurrence of PDAC has been described in individuals with severe and long-standing asthma. Here we explored the potential link between PDAC and the glucocorticoid (GC) budesonide, a first-line therapy to treat asthma., Methods: We tested the effect of budesonide and the classical GCs on the morphology, proliferation, migration and invasiveness of patient-derived PDAC cells and pancreatic cancer cell lines, using 2D and 3D cultures in vitro. Furthermore, a xenograft model was used to investigate the effect of budesonide on PDAC tumor growth in vivo. Finally, we combined genome-wide transcriptome analysis with genetic and pharmacological approaches to explore the mechanisms underlying budesonide activities in the different environmental conditions., Results: We found that in 2D culture settings, high micromolar concentrations of budesonide reduced the mesenchymal invasive/migrating features of PDAC cells, without affecting proliferation or survival. This activity was specific and independent of the Glucocorticoid Receptor (GR). Conversely, in a more physiological 3D environment, low nanomolar concentrations of budesonide strongly reduced PDAC cell proliferation in a GR-dependent manner. Accordingly, we found that budesonide reduced PDAC tumor growth in vivo. Mechanistically, we demonstrated that the 3D environment drives the cells towards a general metabolic reprogramming involving protein, lipid, and energy metabolism (e.g., increased glycolysis dependency). This metabolic change sensitizes PDAC cells to the anti-proliferative effect of budesonide, which instead induces opposite changes (e.g., increased mitochondrial oxidative phosphorylation). Finally, we provide evidence that budesonide inhibits PDAC growth, at least in part, through the tumor suppressor CDKN1C/p57Kip2., Conclusions: Collectively, our study reveals that the microenvironment influences the susceptibility of PDAC cells to GCs and provides unprecedented evidence for the anti-proliferative activity of budesonide on PDAC cells in 3D conditions, in vitro and in vivo. Our findings may explain, at least in part, the reason for the lower occurrence of pancreatic cancer in asthmatic patients and suggest a potential suitability of budesonide for clinical trials as a therapeutic approach to fight pancreatic cancer., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. SERS-based pH-Dependent detection of sulfites in wine by hydrogel nanocomposites.
- Author
-
Yilmaz D, Miranda B, Lonardo E, Rea I, De Stefano L, and De Luca AC
- Subjects
- Gold, Sulfites, Hydrogels, Hydrogen-Ion Concentration, Wine analysis, Metal Nanoparticles chemistry, Biosensing Techniques, Nanocomposites
- Abstract
Sulfur dioxide (SO
2 ) and sulfites are well-known additives in winemaking due to their preservative properties. Although they can prevent oxidation and inhibit microbial growth, they pose health risks and require limitations on their use. Consequently, the total level of SO2 is regulated and several quantification strategies have been proposed. The approved detection methods require the extraction of SO2 by heating and/or acid treatment. Then, iodine or acid/base titrations are conducted for the detection of liberated SO2 . Although these methods can provide sensitive detection of SO2 , they are complex, time-consuming, and require sample preparation steps and skilled operators. Thus, to overcome these disadvantages, an easy-to-use method, involving simple sample preparation steps, and offering high sensitivity and selectivity, is desirable. Herein, we introduce a SERS-based strategy for SO2 detection in liquids using hydrogel nanocomposites. The hydrogels are prepared by poly(ethylene glycol) diacrylate (PEGDA) in the presence of gold nanoparticles (AuNPs), acting as the SERS substrate. The use of hydrogels ensures a homogenous signal distribution and an efficient collection of SO2 , and drying the hydrogels enhances and stabilizes the obtained SO2 signal. The detection strategy is based on the pH-dependent dissociation of SO2 . By adjusting the pH value of wine to 10 through simple dilutions, SO2 can be directly detected in wine, down to 0.4 ppm, well below the regulatory limits. The proposed method allows for sensitive, direct, cost-effective detection of SO2 by eliminating the loss of the gaseous form of the sample and avoids titration-based detection methods., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
13. Revolutionizing Cancer Treatment: Unveiling New Frontiers by Targeting the (Un)Usual Suspects.
- Author
-
Costa V, Giovannetti E, and Lonardo E
- Abstract
This Special Issue includes original articles and reviews on both established and innovative approaches to cancer targeting, showcased at the 29th IGB Workshop titled "Targeting the (un)usual suspects in cancer" "https://29thigbworkshop [...].
- Published
- 2023
- Full Text
- View/download PDF
14. Editorial: Stem cells in pancreatic cancer.
- Author
-
Lonardo E
- Abstract
Competing Interests: The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
15. Quantifying heterogeneity to drug response in cancer-stroma kinetics.
- Author
-
Alemanno F, Cavo M, Delle Cave D, Fachechi A, Rizzo R, D'Amone E, Gigli G, Lonardo E, Barra A, and Del Mercato LL
- Subjects
- Humans, Gemcitabine, Cell Communication, Cell Line, Tumor, Tumor Microenvironment, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology
- Abstract
A crucial challenge in medicine is choosing which drug (or combination) will be the most advantageous for a particular patient. Usually, drug response rates differ substantially, and the reasons for this response unpredictability remain ambiguous. Consequently, it is central to classify features that contribute to the observed drug response variability. Pancreatic cancer is one of the deadliest cancers with limited therapeutic achievements due to the massive presence of stroma that generates an environment that enables tumor growth, metastasis, and drug resistance. To understand the cancer-stroma cross talk within the tumor microenvironment and to develop personalized adjuvant therapies, there is a necessity for effective approaches that offer measurable data to monitor the effect of drugs at the single-cell level. Here, we develop a computational approach, based on cell imaging, that quantifies the cellular cross talk between pancreatic tumor cells (L3.6pl or AsPC1) and pancreatic stellate cells (PSCs), coordinating their kinetics in presence of the chemotherapeutic agent gemcitabine. We report significant heterogeneity in the organization of cellular interactions in response to the drug. For L3.6pl cells, gemcitabine sensibly decreases stroma-stroma interactions but increases stroma-cancer interactions, overall enhancing motility and crowding. In the AsPC1 case, gemcitabine promotes the interactions among tumor cells, but it does not affect stroma-cancer interplay, possibly suggesting a milder effect of the drug on cell dynamics.
- Published
- 2023
- Full Text
- View/download PDF
16. Editorial: Targeting intra- and extracellular signals contributing to cancer stemness and metastasis in aggressive cancers, Volume II.
- Author
-
Caja L, Lonardo E, and Sancho P
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
17. LAMC2 marks a tumor-initiating cell population with an aggressive signature in pancreatic cancer.
- Author
-
Cave DD, Buonaiuto S, Sainz B Jr, Fantuz M, Mangini M, Carrer A, Di Domenico A, Iavazzo TT, Andolfi G, Cortina C, Sevillano M, Heeschen C, Colonna V, Corona M, Cucciardi A, Di Guida M, Batlle E, De Luca A, and Lonardo E
- Subjects
- Humans, Receptor, Transforming Growth Factor-beta Type I, RNA, Small Interfering, Neoplastic Stem Cells metabolism, Transforming Growth Factor beta, RNA, Messenger, Activin Receptors, Cell Movement genetics, Cell Line, Tumor, Laminin genetics, Laminin metabolism, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology
- Abstract
Background: Tumor-initiating cells (TIC), also known as cancer stem cells, are considered a specific subpopulation of cells necessary for cancer initiation and metastasis; however, the mechanisms by which they acquire metastatic traits are not well understood., Methods: LAMC2 transcriptional levels were evaluated using publicly available transcriptome data sets, and LAMC2 immunohistochemistry was performed using a tissue microarray composed of PDAC and normal pancreas tissues. Silencing and tracing of LAMC2 was performed using lentiviral shRNA constructs and CRISPR/Cas9-mediated homologous recombination, respectively. The contribution of LAMC2 to PDAC tumorigenicity was explored in vitro by tumor cell invasion, migration, sphere-forming and organoids assays, and in vivo by tumor growth and metastatic assays. mRNA sequencing was performed to identify key cellular pathways upregulated in LAMC2 expressing cells. Metastatic spreading induced by LAMC2- expressing cells was blocked by pharmacological inhibition of transforming growth factor beta (TGF-β) signaling., Results: We report a LAMC2-expressing cell population, which is endowed with enhanced self-renewal capacity, and is sufficient for tumor initiation and differentiation, and drives metastasis. mRNA profiling of these cells indicates a prominent squamous signature, and differentially activated pathways critical for tumor growth and metastasis, including deregulation of the TGF-β signaling pathway. Treatment with Vactosertib, a new small molecule inhibitor of the TGF-β type I receptor (activin receptor-like kinase-5, ALK5), completely abrogated lung metastasis, primarily originating from LAMC2-expressing cells., Conclusions: We have identified a highly metastatic subpopulation of TICs marked by LAMC2. Strategies aimed at targeting the LAMC2 population may be effective in reducing tumor aggressiveness in PDAC patients. Our results prompt further study of this TIC population in pancreatic cancer and exploration as a potential therapeutic target and/or biomarker., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
18. SERS Quantification of Galunisertib Delivery in Colorectal Cancer Cells by Plasmonic-Assisted Diatomite Nanoparticles.
- Author
-
Managò S, Tramontano C, Delle Cave D, Chianese G, Zito G, De Stefano L, Terracciano M, Lonardo E, De Luca AC, and Rea I
- Subjects
- Diatomaceous Earth, Gold, Humans, Pyrazoles, Quinolines, Tumor Microenvironment, Colorectal Neoplasms drug therapy, Metal Nanoparticles
- Abstract
The small molecule Galunisertib (LY2157299, LY) shows multiple anticancer activities blocking the transforming growth factor-β1 receptor, responsible for the epithelial-to-mesenchymal transition (EMT) by which colorectal cancer (CRC) cells acquire migratory and metastatic capacities. However, frequent dosing of LY can produce highly toxic metabolites. Alternative strategies to reduce drug side effects can rely on nanoscale drug delivery systems that have led to a medical revolution in the treatment of cancer, improving drug efficacy and lowering drug toxicity. Here, a hybrid nanosystem (DNP-AuNPs-LY@Gel) made of a porous diatomite nanoparticle decorated with plasmonic gold nanoparticles, in which LY is retained by a gelatin shell, is proposed. The multifunctional capability of the nanosystem is demonstrated by investigating the efficient LY delivery, the enhanced EMT reversion in CRCs and the intracellular quantification of drug release with a sub-femtogram resolution by surface-enhanced Raman spectroscopy (SERS). The LY release trigger is the pH sensitivity of the gelatin shell to the CRC acidic microenvironment. The drug release is real-time monitored at single-cell level by analyzing the SERS signals of LY in CRC cells. The higher efficiency of LY delivered by the DNP-AuNPs-LY@Gel complex paves the way to an alternative strategy for lowering drug dosing and consequent side effects., (© 2021 The Authors. Small published by Wiley-VCH GmbH.)
- Published
- 2021
- Full Text
- View/download PDF
19. Nodal-induced L1CAM/CXCR4 subpopulation sustains tumor growth and metastasis in colorectal cancer derived organoids.
- Author
-
Cave DD, Hernando-Momblona X, Sevillano M, Minchiotti G, and Lonardo E
- Subjects
- Animals, Cell Line, Tumor, Cell Movement physiology, Colorectal Neoplasms pathology, Humans, Mice, Organoids pathology, Signal Transduction physiology, Tumor Microenvironment physiology, Cell Proliferation physiology, Colorectal Neoplasms metabolism, Neoplasm Metastasis pathology, Neural Cell Adhesion Molecule L1 metabolism, Nodal Protein metabolism, Organoids metabolism, Receptors, CXCR4 metabolism
- Abstract
Background: Colorectal cancer (CRC) is currently the third leading cause for cancer-related mortality. Cancer stem cells have been implicated in colorectal tumor growth, but their specific role in tumor biology, including metastasis, is still uncertain. Methods: Increased expression of L1CAM, CXCR4 and NODAL was identified in tumor section of patients with CRC and in patients-derived-organoids (PDOs). The expression of L1CAM, CXCR4 and NODAL was evaluated using quantitative real-time PCR, western blotting, immunofluorescence, immunohistochemistry and flow cytometry. The effects of the L1CAM, CXCR4 and NODAL on tumor growth, proliferation, migration, invasion, colony-formation ability, metastasis and chemoresistance were investigated both in vitro and in vivo . Results: We found that human colorectal cancer tissue contains cancer stem cells defined by L1CAM
high /CXCR4high expression that is activated by Nodal in hypoxic microenvironment. This L1CAMhigh /CXCR4high population is tumorigenic, highly resistant to standard chemotherapy, and determines the metastatic phenotype of the individual tumor. Depletion of the L1CAMhigh /CXCR4high population drastically reduces the tumorigenic potential and the metastatic phenotype of colorectal tumors. Conclusion: In conclusion, we demonstrated that a subpopulation of migrating L1CAMhigh /CXCR4high is essential for tumor progression. Together, these findings suggest that strategies aimed at modulating the Nodal signaling could have important clinical applications to inhibit colorectal cancer-derived metastasis., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)- Published
- 2021
- Full Text
- View/download PDF
20. The Revolutionary Roads to Study Cell-Cell Interactions in 3D In Vitro Pancreatic Cancer Models.
- Author
-
Delle Cave D, Rizzo R, Sainz B Jr, Gigli G, Del Mercato LL, and Lonardo E
- Abstract
Pancreatic cancer, the fourth most common cancer worldwide, shows a highly unsuccessful therapeutic response. In the last 10 years, neither important advancements nor new therapeutic strategies have significantly impacted patient survival, highlighting the need to pursue new avenues for drug development discovery and design. Advanced cellular models, resembling as much as possible the original in vivo tumor environment, may be more successful in predicting the efficacy of future anti-cancer candidates in clinical trials. In this review, we discuss novel bioengineered platforms for anticancer drug discovery in pancreatic cancer, from traditional two-dimensional models to innovative three-dimensional ones.
- Published
- 2021
- Full Text
- View/download PDF
21. A synergic approach to enhance long-term culture and manipulation of MiaPaCa-2 pancreatic cancer spheroids.
- Author
-
Cavo M, Delle Cave D, D'Amone E, Gigli G, Lonardo E, and Del Mercato LL
- Subjects
- Cell Line, Tumor, Collagen metabolism, Culture Media metabolism, Drug Combinations, Humans, Hydrophobic and Hydrophilic Interactions, Laminin metabolism, Methylcellulose chemistry, Proteoglycans metabolism, Cell Culture Techniques methods, Pancreatic Neoplasms pathology, Spheroids, Cellular pathology
- Abstract
Tumour spheroids have the potential to be used as preclinical chemo-sensitivity assays. However, the production of three-dimensional (3D) tumour spheroids remains challenging as not all tumour cell lines form spheroids with regular morphologies and spheroid transfer often induces disaggregation. In the field of pancreatic cancer, the MiaPaCa-2 cell line is an interesting model for research but it is known for its difficulty to form stable spheroids; also, when formed, spheroids from this cell line are weak and arduous to manage and to harvest for further analyses such as multiple staining and imaging. In this work, we compared different methods (i.e. hanging drop, round-bottom wells and Matrigel embedding, each of them with or without methylcellulose in the media) to evaluate which one allowed to better overpass these limitations. Morphometric analysis indicated that hanging drop in presence of methylcellulose leaded to well-organized spheroids; interestingly, quantitative PCR (qPCR) analysis reflected the morphometric characterization, indicating that same spheroids expressed the highest values of CD44, VIMENTIN, TGF-β1 and Ki-67. In addition, we investigated the generation of MiaPaCa-2 spheroids when cultured on substrates of different hydrophobicity, in order to minimize the area in contact with the culture media and to further improve spheroid formation.
- Published
- 2020
- Full Text
- View/download PDF
22. TGF-β1 secreted by pancreatic stellate cells promotes stemness and tumourigenicity in pancreatic cancer cells through L1CAM downregulation.
- Author
-
Cave DD, Di Guida M, Costa V, Sevillano M, Ferrante L, Heeschen C, Corona M, Cucciardi A, and Lonardo E
- Subjects
- Carcinogenesis genetics, Carcinogenesis pathology, Cell Line, Tumor, Humans, Neoplasm Proteins genetics, Neoplastic Stem Cells pathology, Neural Cell Adhesion Molecule L1 genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Pancreatic Stellate Cells pathology, Transforming Growth Factor beta1 genetics, Carcinogenesis metabolism, Down-Regulation, Gene Expression Regulation, Neoplastic, Neoplasm Proteins metabolism, Neoplastic Stem Cells metabolism, Neural Cell Adhesion Molecule L1 biosynthesis, Pancreatic Neoplasms metabolism, Pancreatic Stellate Cells metabolism, Transforming Growth Factor beta1 metabolism
- Abstract
Pancreatic stellate cells (PSCs) secrete high levels of transforming growth factor-β1 (TGF-β1) that contributes to the development of pancreatic ductal adenocarcinoma (PDAC). TGF-β1 modulates the expression of L1 cell adhesion molecule (L1CAM), but its role in tumour progression still remains controversial. To clarify L1 function in PDAC and cellular phenotypes, we performed L1CAM cell sorting, silencing and overexpression in several primary pancreatic cancer cells. PSCs silenced for TGF-β1 were used for crosstalk experiments. We found that TGF-β1 secreted by PSCs negatively regulates L1CAM expression, through canonical TGF-β-Smad2/3 signalling, leading to a more aggressive PDAC phenotype. Cells with reduced expression of L1CAM harboured enhanced stemness potential and tumourigenicity. Inactivation of TGF-β1 signalling in PSCs strongly reduced the aggressiveness of PDAC cells. Our data provide functional proof and mechanistic insights for the tumour-suppressive function of L1CAM via reducing stemness. Rescuing L1CAM expression in cancer cells through targeting of TGF-β1 reverses stemness and bears the potential to improve the still miserable prognosis of PDAC patients.
- Published
- 2020
- Full Text
- View/download PDF
23. Determinants of metastatic competency in colorectal cancer.
- Author
-
Tauriello DV, Calon A, Lonardo E, and Batlle E
- Subjects
- Animals, Carcinogenesis immunology, Carcinogenesis pathology, Colon immunology, Colon pathology, Colorectal Neoplasms diagnosis, Colorectal Neoplasms immunology, Colorectal Neoplasms therapy, Humans, Immunotherapy methods, Molecular Targeted Therapy methods, Neoplasm Metastasis immunology, Neoplastic Stem Cells immunology, Prognosis, Rectum immunology, Rectum pathology, Colorectal Neoplasms pathology, Neoplasm Metastasis pathology, Neoplastic Stem Cells pathology, Tumor Microenvironment
- Abstract
Colorectal cancer (CRC) is one of the most common cancer types and represents a major therapeutic challenge. Although initial events in colorectal carcinogenesis are relatively well characterized and treatment for early-stage disease has significantly improved over the last decades, the mechanisms underlying metastasis - the main cause of death - remain poorly understood. Correspondingly, no effective therapy is currently available for advanced or metastatic disease. There is increasing evidence that colorectal cancer is hierarchically organized and sustained by cancer stem cells, in concert with various stromal cell types. Here, we review the interplay between cancer stem cells and their microenvironment in promoting metastasis and discuss recent insights relating to both patient prognosis and novel targeted treatment strategies. A better understanding of these topics may aid the prevention or reduction of metastatic burden., (© 2016 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
24. ZFP57 maintains the parent-of-origin-specific expression of the imprinted genes and differentially affects non-imprinted targets in mouse embryonic stem cells.
- Author
-
Riso V, Cammisa M, Kukreja H, Anvar Z, Verde G, Sparago A, Acurzio B, Lad S, Lonardo E, Sankar A, Helin K, Feil R, Fico A, Angelini C, Grimaldi G, and Riccio A
- Subjects
- Animals, Binding Sites genetics, Cell Differentiation genetics, CpG Islands genetics, Epigenesis, Genetic, Genetic Loci, Histones metabolism, Lysine metabolism, Methylation, Mice, Models, Genetic, Gene Expression Regulation, Developmental, Genomic Imprinting, Mouse Embryonic Stem Cells metabolism, Nuclear Proteins metabolism, Repressor Proteins metabolism
- Abstract
ZFP57 is necessary for maintaining repressive epigenetic modifications at Imprinting control regions (ICRs). In mouse embryonic stem cells (ESCs), ZFP57 binds ICRs (ICRBS) and many other loci (non-ICRBS). To address the role of ZFP57 on all its target sites, we performed high-throughput and multi-locus analyses of inbred and hybrid mouse ESC lines carrying different gene knockouts. By using an allele-specific RNA-seq approach, we demonstrate that ZFP57 loss results in derepression of the imprinted allele of multiple genes in the imprinted clusters. We also find marked epigenetic differences between ICRBS and non-ICRBS suggesting that different cis-acting regulatory functions are repressed by ZFP57 at these two classes of target loci. Overall, these data demonstrate that ZFP57 is pivotal to maintain the allele-specific epigenetic modifications of ICRs that in turn are necessary for maintaining the imprinted expression over long distances. At non-ICRBS, ZFP57 inactivation results in acquisition of epigenetic features that are characteristic of poised enhancers, suggesting that another function of ZFP57 in early embryogenesis is to repress cis-acting regulatory elements whose activity is not yet required., (© The Author(s) 2016. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2016
- Full Text
- View/download PDF
25. The miR-17-92 cluster counteracts quiescence and chemoresistance in a distinct subpopulation of pancreatic cancer stem cells.
- Author
-
Cioffi M, Trabulo SM, Sanchez-Ripoll Y, Miranda-Lorenzo I, Lonardo E, Dorado J, Reis Vieira C, Ramirez JC, Hidalgo M, Aicher A, Hahn S, Sainz B Jr, and Heeschen C
- Subjects
- Activins metabolism, Animals, Antimetabolites, Antineoplastic therapeutic use, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Cell Cycle Checkpoints drug effects, Cell Self Renewal, Cell Transformation, Neoplastic, Cyclin-Dependent Kinase Inhibitor p21 genetics, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Cyclin-Dependent Kinase Inhibitor p57 metabolism, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Down-Regulation, Epigenesis, Genetic, Female, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Mice, Mice, Nude, MicroRNAs antagonists & inhibitors, MicroRNAs genetics, Neoplastic Stem Cells drug effects, Nodal Protein metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, RNA, Long Noncoding, Signal Transduction, T-Box Domain Proteins genetics, T-Box Domain Proteins metabolism, Transcriptome, Transforming Growth Factor beta1 metabolism, Gemcitabine, Antimetabolites, Antineoplastic pharmacology, Carcinoma, Pancreatic Ductal metabolism, Deoxycytidine analogs & derivatives, Drug Resistance, Neoplasm genetics, MicroRNAs metabolism, Neoplastic Stem Cells metabolism, Pancreatic Neoplasms metabolism
- Abstract
Objective: Cancer stem cells (CSCs) represent the root of many solid cancers including pancreatic ductal adenocarcinoma, are highly chemoresistant and represent the cellular source for disease relapse. However the mechanisms involved in these processes still need to be fully elucidated. Understanding the mechanisms implicated in chemoresistance and metastasis of pancreatic cancer is critical to improving patient outcomes., Design: Micro-RNA (miRNA) expression analyses were performed to identify functionally defining epigenetic signatures in pancreatic CSC-enriched sphere-derived cells and gemcitabine-resistant pancreatic CSCs., Results: We found the miR-17-92 cluster to be downregulated in chemoresistant CSCs versus non-CSCs and demonstrate its crucial relevance for CSC biology. In particular, overexpression of miR-17-92 reduced CSC self-renewal capacity, in vivo tumourigenicity and chemoresistance by targeting multiple NODAL/ACTIVIN/TGF-β1 signalling cascade members as well as directly inhibiting the downstream targets p21, p57 and TBX3. Overexpression of miR-17-92 translated into increased CSC proliferation and their eventual exhaustion via downregulation of p21 and p57. Finally, the translational impact of our findings could be confirmed in preclinical models for pancreatic cancer., Conclusions: Our findings therefore identify the miR-17-92 cluster as a functionally determining family of miRNAs in CSCs, and highlight the putative potential of developing modulators of this cluster to overcome drug resistance in pancreatic CSCs., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/)
- Published
- 2015
- Full Text
- View/download PDF
26. Studying Pancreatic Cancer Stem Cell Characteristics for Developing New Treatment Strategies.
- Author
-
Lonardo E, Cioffi M, Sancho P, Crusz S, and Heeschen C
- Subjects
- Animals, Cell Culture Techniques, Female, Humans, Metformin pharmacology, Metformin therapeutic use, Mice, Mice, Inbred NOD, Mice, Nude, Mice, SCID, Xenograft Model Antitumor Assays, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal pathology, Neoplastic Stem Cells pathology, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms pathology
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) contains a subset of exclusively tumorigenic cancer stem cells (CSCs) which have been shown to drive tumor initiation, metastasis and resistance to radio- and chemotherapy. Here we describe a specific methodology for culturing primary human pancreatic CSCs as tumor spheres in anchorage-independent conditions. Cells are grown in serum-free, non-adherent conditions in order to enrich for CSCs while their more differentiated progenies do not survive and proliferate during the initial phase following seeding of single cells. This assay can be used to estimate the percentage of CSCs present in a population of tumor cells. Both size (which can range from 35 to 250 micrometers) and number of tumor spheres formed represents CSC activity harbored in either bulk populations of cultured cancer cells or freshly harvested and digested tumors. Using this assay, we recently found that metformin selectively ablates pancreatic CSCs; a finding that was subsequently further corroborated by demonstrating diminished expression of pluripotency-associated genes/surface markers and reduced in vivo tumorigenicity of metformin-treated cells. As the final step for preclinical development we treated mice bearing established tumors with metformin and found significantly prolonged survival. Clinical studies testing the use of metformin in patients with PDAC are currently underway (e.g., NCT01210911, NCT01167738, and NCT01488552). Mechanistically, we found that metformin induces a fatal energy crisis in CSCs by enhancing reactive oxygen species (ROS) production and reducing mitochondrial transmembrane potential. In contrast, non-CSCs were not eliminated by metformin treatment, but rather underwent reversible cell cycle arrest. Therefore, our study serves as a successful example for the potential of in vitro sphere formation as a screening tool to identify compounds that potentially target CSCs, but this technique will require further in vitro and in vivo validation to eliminate false discoveries.
- Published
- 2015
- Full Text
- View/download PDF
27. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer.
- Author
-
Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando-Momblona X, Iglesias M, Sevillano M, Palomo-Ponce S, Tauriello DV, Byrom D, Cortina C, Morral C, Barceló C, Tosi S, Riera A, Attolini CS, Rossell D, Sancho E, and Batlle E
- Subjects
- Animals, Cluster Analysis, Colorectal Neoplasms classification, Colorectal Neoplasms pathology, Fibroblasts pathology, Gene Expression Regulation, Neoplastic, HT29 Cells, Humans, Mice, Mice, Nude, Microarray Analysis, Neoplasm Invasiveness, Neoplasm Metastasis, Neoplastic Stem Cells pathology, Prognosis, Stromal Cells metabolism, Stromal Cells pathology, Transcriptome, Colorectal Neoplasms diagnosis, Colorectal Neoplasms genetics, Fibroblasts metabolism, Neoplastic Stem Cells metabolism
- Abstract
Recent molecular classifications of colorectal cancer (CRC) based on global gene expression profiles have defined subtypes displaying resistance to therapy and poor prognosis. Upon evaluation of these classification systems, we discovered that their predictive power arises from genes expressed by stromal cells rather than epithelial tumor cells. Bioinformatic and immunohistochemical analyses identify stromal markers that associate robustly with disease relapse across the various classifications. Functional studies indicate that cancer-associated fibroblasts (CAFs) increase the frequency of tumor-initiating cells, an effect that is dramatically enhanced by transforming growth factor (TGF)-β signaling. Likewise, we find that all poor-prognosis CRC subtypes share a gene program induced by TGF-β in tumor stromal cells. Using patient-derived tumor organoids and xenografts, we show that the use of TGF-β signaling inhibitors to block the cross-talk between cancer cells and the microenvironment halts disease progression.
- Published
- 2015
- Full Text
- View/download PDF
28. Intracellular autofluorescence: a biomarker for epithelial cancer stem cells.
- Author
-
Miranda-Lorenzo I, Dorado J, Lonardo E, Alcala S, Serrano AG, Clausell-Tormos J, Cioffi M, Megias D, Zagorac S, Balic A, Hidalgo M, Erkan M, Kleeff J, Scarpa A, Sainz B Jr, and Heeschen C
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2, ATP-Binding Cassette Transporters metabolism, Animals, Autophagy, Autophagy-Related Protein 12, Carcinoma, Hepatocellular pathology, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Pancreatic Ductal pathology, Colorectal Neoplasms pathology, Female, Humans, Liver Neoplasms pathology, Lung Neoplasms pathology, Mice, Mice, Nude, Microtubule-Associated Proteins biosynthesis, Neoplasm Proteins metabolism, Pancreatic Neoplasms pathology, Small Ubiquitin-Related Modifier Proteins biosynthesis, Tumor Cells, Cultured, Biomarkers, Tumor metabolism, Cell Separation methods, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Optical Imaging methods, Riboflavin metabolism
- Abstract
Cancer stem cells (CSCs) are thought to drive tumor growth, metastasis and chemoresistance. Although surface markers such as CD133 and CD44 have been successfully used to isolate CSCs, their expression is not exclusively linked to the CSC phenotype and is prone to environmental alteration. We identified cells with an autofluorescent subcellular compartment that exclusively showed CSC features across different human tumor types. Primary tumor-derived autofluorescent cells did not overlap with side-population (SP) cells, were enriched in sphere culture and during chemotherapy, strongly expressed pluripotency-associated genes, were highly metastatic and showed long-term in vivo tumorigenicity, even at the single-cell level. Autofluorescence was due to riboflavin accumulation in membrane-bounded cytoplasmic structures bearing ATP-dependent ABCG2 transporters. In summary, we identified and characterized an intrinsic autofluorescent phenotype in CSCs of diverse epithelial cancers and used this marker to isolate and characterize these cells.
- Published
- 2014
- Full Text
- View/download PDF
29. Metformin targets the metabolic achilles heel of human pancreatic cancer stem cells.
- Author
-
Lonardo E, Cioffi M, Sancho P, Sanchez-Ripoll Y, Trabulo SM, Dorado J, Balic A, Hidalgo M, and Heeschen C
- Subjects
- Biomarkers metabolism, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Cell Transformation, Neoplastic drug effects, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Disease Progression, Gene Expression Regulation, Neoplastic drug effects, Humans, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Stromal Cells drug effects, Stromal Cells metabolism, Metformin pharmacology, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Pancreatic Neoplasms metabolism
- Abstract
Pancreatic ductal adenocarcinomas contain a subset of exclusively tumorigenic cancer stem cells (CSCs), which are capable of repopulating the entire heterogeneous cancer cell populations and are highly resistant to standard chemotherapy. Here we demonstrate that metformin selectively ablated pancreatic CSCs as evidenced by diminished expression of pluripotency-associated genes and CSC-associated surface markers. Subsequently, the ability of metformin-treated CSCs to clonally expand in vitro was irreversibly abrogated by inducing apoptosis. In contrast, non-CSCs preferentially responded by cell cycle arrest, but were not eliminated by metformin treatment. Mechanistically, metformin increased reactive oxygen species production in CSC and reduced their mitochondrial transmembrane potential. The subsequent induction of lethal energy crisis in CSCs was independent of AMPK/mTOR. Finally, in primary cancer tissue xenograft models metformin effectively reduced tumor burden and prevented disease progression; if combined with a stroma-targeting smoothened inhibitor for enhanced tissue penetration, while gemcitabine actually appeared dispensable.
- Published
- 2013
- Full Text
- View/download PDF
30. Cripto regulates skeletal muscle regeneration and modulates satellite cell determination by antagonizing myostatin.
- Author
-
Guardiola O, Lafuste P, Brunelli S, Iaconis S, Touvier T, Mourikis P, De Bock K, Lonardo E, Andolfi G, Bouché A, Liguori GL, Shen MM, Tajbakhsh S, Cossu G, Carmeliet P, and Minchiotti G
- Subjects
- Aging metabolism, Animals, Cell Proliferation, Gene Deletion, Gene Targeting, Hypertrophy, Mice, Mice, Inbred C57BL, Models, Animal, Muscle Development, Muscle Fibers, Skeletal metabolism, Muscle Fibers, Skeletal pathology, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Myoblasts metabolism, Myoblasts pathology, Myostatin metabolism, Signal Transduction, Cell Lineage, Epidermal Growth Factor metabolism, Membrane Glycoproteins metabolism, Muscle, Skeletal physiology, Myostatin antagonists & inhibitors, Neoplasm Proteins metabolism, Regeneration, Satellite Cells, Skeletal Muscle metabolism, Satellite Cells, Skeletal Muscle pathology
- Abstract
Skeletal muscle regeneration mainly depends on satellite cells, a population of resident muscle stem cells. However, our understanding of the molecular mechanisms underlying satellite cell activation is still largely undefined. Here, we show that Cripto, a regulator of early embryogenesis, is a novel regulator of muscle regeneration and satellite cell progression toward the myogenic lineage. Conditional inactivation of cripto in adult satellite cells compromises skeletal muscle regeneration, whereas gain of function of Cripto accelerates regeneration, leading to muscle hypertrophy. Moreover, we provide evidence that Cripto modulates myogenic cell determination and promotes proliferation by antagonizing the TGF-β ligand myostatin. Our data provide unique insights into the molecular and cellular basis of Cripto activity in skeletal muscle regeneration and raise previously undescribed implications for stem cell biology and regenerative medicine.
- Published
- 2012
- Full Text
- View/download PDF
31. Pancreatic stellate cells form a niche for cancer stem cells and promote their self-renewal and invasiveness.
- Author
-
Lonardo E, Frias-Aldeguer J, Hermann PC, and Heeschen C
- Subjects
- Activin Receptors, Type I metabolism, Carcinoma, Pancreatic Ductal drug therapy, Cell Line, Tumor, Fibrosis, Hedgehog Proteins antagonists & inhibitors, Hedgehog Proteins metabolism, Humans, Neoplasm Invasiveness, Neoplastic Stem Cells pathology, Pancreas pathology, Pancreatic Neoplasms drug therapy, Pancreatitis, Chronic pathology, Stem Cell Niche drug effects, Tumor Microenvironment, Carcinoma, Pancreatic Ductal pathology, Neoplastic Stem Cells physiology, Pancreatic Neoplasms pathology, Pancreatic Stellate Cells physiology, Stem Cell Niche physiology
- Abstract
Chronic pancreatitis and pancreatic ductal adenocarcinoma (PDAC) are characterized by extensive fibrosis. Importantly, in PDAC, this results in poor vascularization and impaired drug delivery to the cancer cells. Therefore, the combined targeting of pancreatic tumor stroma and chemotherapy should enhance response rates, but the negative outcome of a recent phase III clinical trial for the combination of chemotherapy and hedgehog pathway inhibition suggests that other means also need to be considered. Emerging data indicate that elimination of cancer stem cells as the root of the cancer is of pivotal importance for efficient treatment of pancreatic cancer. Recently, we demonstrated in a highly relevant preclinical mouse model for primary pancreatic cancers that the combination of cancer stem cell-targeting strategies in combination with a stroma-targeting agent, such as a hedgehog pathway inhibitor and chemotherapy, results in significantly enhanced long-term and progression-free survival. In the present study, we demonstrate mechanistically that Nodal-expressing pancreatic stellate cells are an important component of the tumor stroma for creating a paracrine niche for pancreatic cancer stem cells. Secretion of the embryonic morphogens Nodal/Activin by pancreatic stellate cells promoted in vitro sphere formation and invasiveness of pancreatic cancer stem cells in an Alk4-dependent manner. These data imply that the pancreatic cancer stem cell phenotype is promoted by paracrine Nodal/Activin signaling at the tumor-stroma interface. Therefore, targeting the tumor microenvironment is not only able to improve drug delivery but, even more importantly, destroys the cancer stem cell niche and, therefore, should be an integral part of cancer stem cell-based treatment strategies.
- Published
- 2012
- Full Text
- View/download PDF
32. Nodal/Activin signaling drives self-renewal and tumorigenicity of pancreatic cancer stem cells and provides a target for combined drug therapy.
- Author
-
Lonardo E, Hermann PC, Mueller MT, Huber S, Balic A, Miranda-Lorenzo I, Zagorac S, Alcala S, Rodriguez-Arabaolaza I, Ramirez JC, Torres-Ruíz R, Garcia E, Hidalgo M, Cebrián DÁ, Heuchel R, Löhr M, Berger F, Bartenstein P, Aicher A, and Heeschen C
- Subjects
- AC133 Antigen, Animals, Antigens, CD metabolism, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor metabolism, Cell Proliferation, Cell Transformation, Neoplastic metabolism, Female, Gene Targeting, Glycoproteins metabolism, Humans, Mice, Mice, Nude, Neoplastic Stem Cells metabolism, Pancreatic Neoplasms metabolism, Peptides metabolism, Pluripotent Stem Cells metabolism, Spheroids, Cellular metabolism, Spheroids, Cellular pathology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Activins metabolism, Cell Transformation, Neoplastic pathology, Neoplastic Stem Cells pathology, Nodal Protein metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms pathology, Signal Transduction
- Abstract
Nodal and Activin belong to the TGF-β superfamily and are important regulators of embryonic stem cell fate. Here we investigated whether Nodal and Activin regulate self-renewal of pancreatic cancer stem cells. Nodal and Activin were hardly detectable in more differentiated pancreatic cancer cells, while cancer stem cells and stroma-derived pancreatic stellate cells markedly overexpressed Nodal and Activin, but not TGF-β. Knockdown or pharmacological inhibition of the Nodal/Activin receptor Alk4/7 in cancer stem cells virtually abrogated their self-renewal capacity and in vivo tumorigenicity, and reversed the resistance of orthotopically engrafted cancer stem cells to gemcitabine. However, engrafted primary human pancreatic cancer tissue with a substantial stroma showed no response due to limited drug delivery. The addition of a stroma-targeting hedgehog pathway inhibitor enhanced delivery of the Nodal/Activin inhibitor and translated into long-term, progression-free survival. Therefore, inhibition of the Alk4/7 pathway, if combined with hedgehog pathway inhibition and gemcitabine, provides a therapeutic strategy for targeting cancer stem cells., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
33. Pancreatic cancer stem cells: new insights and perspectives.
- Author
-
Dorado J, Lonardo E, Miranda-Lorenzo I, and Heeschen C
- Subjects
- Biomarkers, Tumor metabolism, Carcinoma, Pancreatic Ductal therapy, Humans, Models, Biological, Neoplastic Stem Cells metabolism, Pancreatic Neoplasms therapy, Carcinoma, Pancreatic Ductal pathology, Neoplastic Stem Cells pathology, Pancreatic Neoplasms pathology
- Abstract
Since the identification of self-renewing cells in the hematopoietic system several decades ago, stem cells have changed the way we study biology and medicine. Solid tumors contain a distinct subpopulation of cells that have stem cell characteristics and are exclusively responsible for tumorigenicity. This discovery has led to the development of the stem cell concept of cancer, which proposes that a subpopulation of self-renewing tumor cells, also termed cancer stem cells, is responsible for tumorigenesis and metastasis. This contrasts with the stochastic model of tumor development, which holds that all tumor cells are capable of tumor initiation. Different subpopulations of cancer stem cells have been identified in pancreatic ductal adenocarcinoma, based on the use of combinations of surface markers that allow their isolation, propagation, and further characterization. Importantly, cancer stem cells are not only capable of self-renewal and differentiation, but may also confer virulence via immune system evasion and multidrug resistance, and potentially via vasculogenic mimicry and transition to migratory and metastasizing derivatives. Therapeutic targeting of this subset of cells and the pathways defining their virulence holds great promise for the development of more effective strategies for the amelioration and eradication of this most lethal form of cancer.
- Published
- 2011
- Full Text
- View/download PDF
34. Pancreatic cancer stem cells - update and future perspectives.
- Author
-
Lonardo E, Hermann PC, and Heeschen C
- Subjects
- Animals, Humans, Mice, Pancreatic Neoplasms pathology, Stem Cells pathology
- Abstract
Solid tumours are the most common cancers and represent a major therapeutic challenge. The cancer stem cell hypothesis is an attractive model to explain the functional heterogeneity commonly observed in solid tumours. It proposes a hierarchical organization of tumours, in which a subpopulation of stem cell-like cells sustains tumour growth, metastasis, and resistance to therapy. We will present the most recent advances in the cancer stem cell field, with particular emphasis on pancreatic cancer as one of the deadliest human tumours, and highlight open questions and caveats to be addressed in future studies. There is increasing evidence that solid tumours including pancreatic cancer are hierarchically organized and sustained by a distinct subpopulation of cancer stem cells. However, direct evidence for the validity of the cancer stem cell hypothesis in human pancreatic cancer remains controversial due to the limitations of xenograft models but supportive data are now emerging from mouse models using related or different sets of markers for the identification of murine cancer stem cells. Therefore, while the clinical relevance of cancer stem cells remains a fundamental issue for this rapidly emerging field, current findings clearly suggest that specific elimination of these cells is possible and therapeutically relevant. Targeting of signalling pathways that are of particular importance for the maintenance and the elimination of cancer stem cell as the proposed root of the tumour may lead to the development of novel treatment regimens for pancreatic cancer. Here we will review the current literature on pancreatic cancer stem cells and the future perspective of this rapidly emerging field., (Copyright © 2010 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
35. A small synthetic cripto blocking Peptide improves neural induction, dopaminergic differentiation, and functional integration of mouse embryonic stem cells in a rat model of Parkinson's disease.
- Author
-
Lonardo E, Parish CL, Ponticelli S, Marasco D, Ribeiro D, Ruvo M, De Falco S, Arenas E, and Minchiotti G
- Subjects
- Activin Receptors, Type I metabolism, Animals, Disease Models, Animal, Embryonic Stem Cells drug effects, Epidermal Growth Factor metabolism, Membrane Glycoproteins metabolism, Mice, Neoplasm Proteins metabolism, Neurons cytology, Parkinson Disease therapy, Protein Binding drug effects, Rats, Reverse Transcriptase Polymerase Chain Reaction, Stem Cell Transplantation, Cell Differentiation drug effects, Embryonic Stem Cells cytology, Neurons drug effects, Oligopeptides pharmacology, Oligopeptides therapeutic use, Parkinson Disease drug therapy
- Abstract
Cripto is a glycosylphosphatidylinositol-anchored coreceptor that binds Nodal and the activin type I (ALK)-4 receptor, and is involved in cardiac differentiation of mouse embryonic stem cells (mESCs). Interestingly, genetic ablation of cripto results in increased neuralization and midbrain dopaminergic (DA) differentiation of mESCs, as well as improved DA cell replacement therapy (CRT) in a model of Parkinson's disease (PD). In this study, we developed a Cripto specific blocking tool that would mimic the deletion of cripto, but could be easily applied to embryonic stem cell (ESC) lines without the need of genetic manipulation. We thus screened a combinatorial peptide library and identified a tetrameric tripeptide, Cripto blocking peptide (BP), which prevents Cripto/ALK-4 receptor interaction and interferes with Cripto signaling. Cripto BP treatment favored neuroectoderm formation and promoted midbrain DA neuron differentiation of mESCs in vitro and in vivo. Remarkably, Cripto BP-treated ESCs, when transplanted into the striatum of PD rats, enhanced functional recovery and reduced tumor formation, mimicking the effect of genetic ablation of cripto. We therefore suggest that specific blockers such as Cripto BP may be used to improve the differentiation of ESC-derived DA neurons in vitro and their engraftment in vivo, bringing us closer towards an application of ESCs in CRT.
- Published
- 2010
- Full Text
- View/download PDF
36. Lack of sik1 in mouse embryonic stem cells impairs cardiomyogenesis by down-regulating the cyclin-dependent kinase inhibitor p57kip2.
- Author
-
Romito A, Lonardo E, Roma G, Minchiotti G, Ballabio A, and Cobellis G
- Subjects
- Animals, Blotting, Western, Cell Cycle, Cell Differentiation, Cell Line, Cell Proliferation, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p57 genetics, Down-Regulation, Embryonic Stem Cells cytology, Flow Cytometry, Gene Expression Profiling, Humans, Mice, Mice, Knockout, Myocytes, Cardiac cytology, Neurons cytology, Neurons metabolism, Oligonucleotide Array Sequence Analysis, Protein Serine-Threonine Kinases genetics, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Cyclin-Dependent Kinase Inhibitor p57 metabolism, Embryonic Stem Cells metabolism, Myocytes, Cardiac metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Sik1 (salt inducible kinase 1) is a serine/threonine kinase that belongs to the stress- and energy-sensing AMP-activated protein kinase family. During murine embryogenesis, sik1 marks the monolayer of future myocardial cells that will populate first the primitive ventricle, and later the primitive atrium suggesting its involvement in cardiac cell differentiation and/or heart development. Despite that observation, the involvement of sik1 in cardiac differentiation is still unknown. We examined the sik1 function during cardiomyocyte differentiation using the ES-derived embryoid bodies. We produced a null embryonic stem cell using a gene-trap cell line carrying an insertion in the sik1 locus. In absence of the sik1 protein, the temporal appearance of cardiomyocytes is delayed. Expression profile analysis revealed sik1 as part of a genetic network that controls the cell cycle, where the cyclin-dependent kinase inhibitor p57(Kip2) is directly involved. Collectively, we provided evidence that sik1-mediated effects are specific for cardiomyogenesis regulating cardiomyoblast cell cycle exit toward terminal differentiation.
- Published
- 2010
- Full Text
- View/download PDF
37. Cripto-1 is required for hypoxia to induce cardiac differentiation of mouse embryonic stem cells.
- Author
-
Bianco C, Cotten C, Lonardo E, Strizzi L, Baraty C, Mancino M, Gonzales M, Watanabe K, Nagaoka T, Berry C, Arai AE, Minchiotti G, and Salomon DS
- Subjects
- Animals, Biomarkers metabolism, Cell Line, Embryonic Stem Cells cytology, Epidermal Growth Factor genetics, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Membrane Glycoproteins genetics, Mice, Mice, Knockout, Myocardium cytology, Myocardium metabolism, Myocytes, Cardiac cytology, Neoplasm Proteins genetics, Promoter Regions, Genetic, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Response Elements, Signal Transduction physiology, Swine, Cell Differentiation physiology, Embryonic Stem Cells physiology, Epidermal Growth Factor metabolism, Heart anatomy & histology, Heart embryology, Hypoxia metabolism, Membrane Glycoproteins metabolism, Myocytes, Cardiac physiology, Neoplasm Proteins metabolism
- Abstract
Cripto-1 is a membrane-bound protein that is highly expressed in embryonic stem cells and in human tumors. In the present study, we investigated the effect of low levels of oxygen, which occurs naturally in rapidly growing tissues, on Cripto-1 expression in mouse embryonic stem (mES) cells and in human embryonal carcinoma cells. During hypoxia, Cripto-1 expression levels were significantly elevated in mES cells and in Ntera-2 or NCCIT human embryonal carcinoma cells, as compared with cells growing with normal oxygen levels. The transcription factor hypoxia-inducible factor-1alpha directly regulated Cripto-1 expression by binding to hypoxia-responsive elements within the promoter of mouse and human Cripto-1 genes in mES and NCCIT cells, respectively. Furthermore, hypoxia modulated differentiation of mES cells by enhancing formation of beating cardiomyocytes as compared with mES cells that were differentiated under normoxia. However, hypoxia failed to induce differentiation of mES cells into cardiomyocytes in the absence of Cripto-1 expression, demonstrating that Cripto-1 is required for hypoxia to fully differentiate mES cells into cardiomyocytes. Finally, cardiac tissue samples derived from patients who had suffered ischemic heart disease showed a dramatic increase in Cripto-1 expression as compared with nonischemic heart tissue samples, suggesting that hypoxia may also regulate Cripto-1 in vivo.
- Published
- 2009
- Full Text
- View/download PDF
38. G protein-coupled receptor APJ and its ligand apelin act downstream of Cripto to specify embryonic stem cells toward the cardiac lineage through extracellular signal-regulated kinase/p70S6 kinase signaling pathway.
- Author
-
D'Aniello C, Lonardo E, Iaconis S, Guardiola O, Liguoro AM, Liguori GL, Autiero M, Carmeliet P, and Minchiotti G
- Subjects
- Adipokines, Animals, Apelin, Apelin Receptors, Cell Line, Cells, Cultured, Embryo, Mammalian metabolism, Embryonic Stem Cells metabolism, Epidermal Growth Factor genetics, GTP-Binding Proteins metabolism, Intercellular Signaling Peptides and Proteins, Membrane Glycoproteins genetics, Mice, Mice, Knockout, Myocardium cytology, Myocardium metabolism, Myocytes, Cardiac metabolism, Neoplasm Proteins genetics, Signal Transduction physiology, Smad2 Protein metabolism, Carrier Proteins metabolism, Cell Differentiation physiology, Embryonic Stem Cells cytology, Epidermal Growth Factor metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Membrane Glycoproteins metabolism, Myocytes, Cardiac cytology, Neoplasm Proteins metabolism, Receptors, G-Protein-Coupled metabolism, Ribosomal Protein S6 Kinases, 70-kDa metabolism
- Abstract
Rationale: Pluripotent stem cells represent a powerful model system to study the early steps of cardiac specification for which the molecular control is largely unknown. The EGF-CFC (epidermal growth factor-Cripto/FRL-1/Cryptic) Cripto protein is essential for cardiac myogenesis in embryonic stem cells (ESCs)., Objective: Here, we study the role of apelin and its G protein-coupled receptor, APJ, as downstream targets of Cripto both in vivo and in ESC differentiation., Methods and Results: Gain-of-function experiments show that APJ suppresses neuronal differentiation and restores the cardiac program in Cripto(-/-) ESCs. Loss-of-function experiments point for a central role for APJ/apelin in the gene regulatory cascade promoting cardiac specification and differentiation in ESCs. Remarkably, we show for the first time that apelin promotes mammalian cardiomyogenesis via activation of mitogen-activated protein kinase/p70S6 through coupling to a Go/Gi protein., Conclusions: Together our data provide evidence for a previously unrecognized function of APJ/apelin in the Cripto signaling pathway governing mesoderm patterning and cardiac specification in mammals.
- Published
- 2009
- Full Text
- View/download PDF
39. Effects of indole-3-acetic acid on Sinorhizobium meliloti survival and on symbiotic nitrogen fixation and stem dry weight production.
- Author
-
Imperlini E, Bianco C, Lonardo E, Camerini S, Cermola M, Moschetti G, and Defez R
- Subjects
- Acetyl Coenzyme A, Bacterial Proteins metabolism, Citrate (si)-Synthase metabolism, Hydroxybutyrates metabolism, Ketoglutarate Dehydrogenase Complex metabolism, Plant Stems growth & development, Polyesters metabolism, Biomass, Indoleacetic Acids metabolism, Medicago truncatula growth & development, Microbial Viability, Nitrogen Fixation drug effects, Plant Growth Regulators metabolism, Sinorhizobium meliloti drug effects
- Abstract
We evaluated the effects of the main auxin phytohormone, indole-3-acetic acid (IAA), on the central metabolism of Sinorhizobium meliloti 1021. We either treated S. meliloti 1021 wild-type cells with 0.5 mM IAA, 1021+, or use a derivative, RD64, of the same strain harboring an additional pathway for IAA biosynthesis (converting tryptophan into IAA via indoleacetamide). We assayed the activity of tricarboxylic acid cycle (TCA) key enzymes and found that activity of citrate synthase and alpha-ketoglutarate dehydrogenase were increased in both 1021+ and RD64 as compared to the wild-type strain. We also showed that the intracellular acetyl-CoA content was enhanced in both RD64 and 1021+ strains when compared to the control strain. The activity of key enzymes, utilizing acetyl-CoA for poly-beta-hydroxybutyrate (PHB) biosynthesis, was also induced. The PHB level measured in these cells were significantly higher than that found in control cells. Moreover, 4-week-long survival experiments showed that 80% of 1021 cells died, whereas 50% of RD64 cells were viable. Medicago truncatula plants nodulated by RD64 (Mt-RD64) showed an induction of both acetylene reduction activity and stem dry weight production.
- Published
- 2009
- Full Text
- View/download PDF
40. Qualitative and quantitative proteomic profiling of cripto(-/-) embryonic stem cells by means of accurate mass LC-MS analysis.
- Author
-
Chambery A, Vissers JP, Langridge JI, Lonardo E, Minchiotti G, Ruvo M, and Parente A
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, Embryonic Stem Cells cytology, Mice, Mice, Knockout, Molecular Sequence Data, Neurons physiology, Proteome analysis, Reproducibility of Results, Chromatography, Liquid methods, Embryonic Stem Cells chemistry, Embryonic Stem Cells physiology, Epidermal Growth Factor genetics, Epidermal Growth Factor metabolism, Mass Spectrometry methods, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Protein Array Analysis methods
- Abstract
Cripto is one of the key regulators of embryonic stem cells (ESCs) differentiation into cardiomyocites vs neuronal fate. Cripto(-/-) murine ESCs have been utilized to investigate the molecular mechanisms underlying early events of mammalian lineage differentiation. 2D/LC-MS/MS and a label-free LC-MS approaches were used to qualitatively and quantitatively profile the cripto(-/-) ESC proteome, providing an integral view of the alterations induced in stem cell functions by deleting the cripto gene.
- Published
- 2009
- Full Text
- View/download PDF
41. Introduction of a novel pathway for IAA biosynthesis to rhizobia alters vetch root nodule development.
- Author
-
Camerini S, Senatore B, Lonardo E, Imperlini E, Bianco C, Moschetti G, Rotino GL, Campion B, and Defez R
- Subjects
- Acetylene metabolism, Agrobacterium tumefaciens genetics, Bacterial Proteins genetics, DNA, Bacterial genetics, Genes, Bacterial, Nitrogen Fixation, Plant Growth Regulators metabolism, Plasmids, Promoter Regions, Genetic, Pseudomonas genetics, Recombinant Proteins genetics, Root Nodules, Plant microbiology, Symbiosis, Vicia growth & development, Indoleacetic Acids metabolism, Rhizobium leguminosarum genetics, Rhizobium leguminosarum metabolism, Root Nodules, Plant growth & development, Vicia microbiology
- Abstract
We introduced into Rhizobium leguminosarum bv. viciae LPR1105 a new pathway for the biosynthesis of the auxin, indole-3-acetic acid (IAA), under the control of a stationary phase-activated promoter active both in free-living bacteria and bacteroids. The newly introduced genes are the iaaM gene from Pseudomonas savastanoi and the tms2 gene from Agrobacterium tumefaciens. Free-living bacteria harbouring the promoter-iaaMtms2 construct release into the growth medium 14-fold more IAA than the wild-type parental strain. This IAA overproducing R. l. viciae, the RD20 strain, elicits the development of vetch root nodules containing up to 60-fold more IAA than nodules infected by the wild-type strain LPR1105. Vetch root nodules derived from RD20 are fewer in number per plant, heavier in terms of dry weight and show an enlarged and more active meristem. A significant increase in acetylene reduction activity was measured in nodules elicited in vetch by RD20.
- Published
- 2008
- Full Text
- View/download PDF
42. Cripto promotes A-P axis specification independently of its stimulatory effect on Nodal autoinduction.
- Author
-
D'Andrea D, Liguori GL, Le Good JA, Lonardo E, Andersson O, Constam DB, Persico MG, and Minchiotti G
- Subjects
- Activin Receptors, Type I genetics, Activin Receptors, Type I metabolism, Animals, Animals, Genetically Modified, Chimera, Epidermal Growth Factor genetics, Feedback, Physiological genetics, Female, Gastrula embryology, Gastrula metabolism, Gene Expression Regulation genetics, Gene Expression Regulation, Developmental genetics, Male, Membrane Glycoproteins genetics, Mice, Mice, Inbred C57BL, Mutation genetics, Neoplasm Proteins genetics, Nodal Protein, Signal Transduction genetics, Smad2 Protein genetics, Smad2 Protein metabolism, Transforming Growth Factor beta genetics, Body Patterning physiology, Embryonic Development physiology, Epidermal Growth Factor metabolism, Membrane Glycoproteins metabolism, Neoplasm Proteins metabolism, Transforming Growth Factor beta metabolism
- Abstract
The EGF-CFC gene cripto governs anterior-posterior (A-P) axis specification in the vertebrate embryo. Existing models suggest that Cripto facilitates binding of Nodal to an ActRII-activin-like kinase (ALK) 4 receptor complex. Cripto also has a crucial function in cellular transformation that is independent of Nodal and ALK4. However, how ALK4-independent Cripto pathways function in vivo has remained unclear. We have generated cripto mutants carrying the amino acid substitution F78A, which blocks the Nodal-ALK4-Smad2 signaling both in embryonic stem cells and cell-based assays. In cripto(F78A/F78A) mouse embryos, Nodal fails to expand its own expression domain and that of cripto, indicating that F78 is essential in vivo to stimulate Smad-dependent Nodal autoinduction. In sharp contrast to cripto-null mutants, cripto(F78A/F78A) embryos establish an A-P axis and initiate gastrulation movements. Our findings provide in vivo evidence that Cripto is required in the Nodal-Smad2 pathway to activate an autoinductive feedback loop, whereas it can promote A-P axis formation and initiate gastrulation movements independently of its stimulatory effect on the canonical Nodal-ALK4-Smad2 signaling pathway.
- Published
- 2008
- Full Text
- View/download PDF
43. [Myocardial T1-201 scintigraphy in the study of the development of acute myocardial infarction. Evaluation of the effects of a drug with metabolic action: creatine phosphate].
- Author
-
Pedone V, Corbelli C, Frondini C, Franchi R, Luppi M, Lonardo E, Balboni A, Monetti N, and di Biase G
- Subjects
- Adult, Aged, Humans, Infant, Newborn, Middle Aged, Myocardial Infarction drug therapy, Myocardial Infarction metabolism, Phosphocreatine metabolism, Phosphocreatine therapeutic use, Radionuclide Imaging, Myocardial Infarction diagnostic imaging, Radioisotopes, Thallium
- Published
- 1984
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.