1
|
Samimi A, Khodayar MJ, Alidadi H, Khodadi E. The Dual Role of ROS in Hematological Malignancies: Stem Cell Protection and Cancer Cell Metastasis. Stem Cell Rev Rep 2021; 16:262-275. [PMID: 31912368 DOI: 10.1007/s12015-019-09949-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND OBJECTIVE Reactive oxygen species (ROS) play crucial role in hematopoiesis, regulation of differentiation, self-renewal, and the balance between quiescence and proliferation of hematopoietic stem cells (HSCs). The HSCs are a small population of undifferentiated cells that reside in the bone marrow (BM) and can undergo self-renewal by giving rise to mature cells. METHODS Relevant literature was identified through a PubMed search (2000-2019) of English-language papers using the following terms: reactive oxygen species, hematopoietic stem cell, leukemic stem cell, leukemia and chemotherapy. RESULTS HSCs are very sensitive to high levels of ROS and increased production of ROS have been attributed to HSC aging. HSC aging induced by both cell intrinsic and extrinsic factors is linked to impaired HSC self-renewal and regeneration. In addition, the elevated ROS levels might even trigger differentiation of Leukemic stem cells (LSCs) and ROS may be involved in the initiation and progression of hematological malignancies, such as leukemia. CONCLUSION Targeting genes involved in ROS in LSCs and HSCs are increasingly being used as a critical target for therapeutic interventions. Appropriate concentration of ROS may be an optimal therapeutic target for treatment of leukemia during chemotherapy, but still more studies are required to better understanding of the of ROS role in blood disorders.
Collapse
Affiliation(s)
- Azin Samimi
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Legal Medicine Organization, Legal Medicine Research Center, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadis Alidadi
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elahe Khodadi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
2
|
Wang L, Li H, Shen X, Zeng J, Yue L, Lin J, Yang J, Zou W, Li Y, Qin D, Wu A, Wu J. Elucidation of the molecular mechanism of Sanguisorba Officinalis L. against leukopenia based on network pharmacology. Biomed Pharmacother 2020; 132:110934. [DOI: 10.1016/j.biopha.2020.110934] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 01/07/2023] Open
|
3
|
Ikonomi N, Kühlwein SD, Schwab JD, Kestler HA. Awakening the HSC: Dynamic Modeling of HSC Maintenance Unravels Regulation of the TP53 Pathway and Quiescence. Front Physiol 2020; 11:848. [PMID: 32848827 PMCID: PMC7411231 DOI: 10.3389/fphys.2020.00848] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/24/2020] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem cells (HSCs) provide all types of blood cells during the entire life of the organism. HSCs are mainly quiescent and can eventually enter the cell cycle to differentiate. HSCs are maintained and tightly regulated in a particular environment. The stem cell niche regulates dormancy and awakening. Deregulations of this interplay can lead to hematopoietic failure and diseases. In this paper, we present a Boolean network model that recapitulates HSC regulation in virtue of external signals coming from the niche. This Boolean network integrates and summarizes the current knowledge of HSC regulation and is based on extensive literature research. Furthermore, dynamic simulations suggest a novel systemic regulation of TP53 in homeostasis. Thereby, our model indicates that TP53 activity is balanced depending on external stimulations, engaging a regulatory mechanism involving ROS regulators and RAS activated transcription factors. Finally, we investigated different mouse models and compared them to in silico knockout simulations. Here, the model could recapitulate in vivo observed behaviors and thus sustains our results.
Collapse
Affiliation(s)
- Nensi Ikonomi
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Silke D Kühlwein
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Julian D Schwab
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| |
Collapse
|
4
|
Network Pharmacology-Based Investigation of the System-Level Molecular Mechanisms of the Hematopoietic Activity of Samul-Tang, a Traditional Korean Herbal Formula. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9048089. [PMID: 32104198 PMCID: PMC7040423 DOI: 10.1155/2020/9048089] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/03/2020] [Indexed: 12/12/2022]
Abstract
Hematopoiesis is a dynamic process of the continuous production of diverse blood cell types to meet the body's physiological demands and involves complex regulation of multiple cellular mechanisms in hematopoietic stem cells, including proliferation, self-renewal, differentiation, and apoptosis. Disruption of the hematopoietic system is known to cause various hematological disorders such as myelosuppression. There is growing evidence on the beneficial effects of herbal medicines on hematopoiesis; however, their mechanism of action remains unclear. In this study, we conducted a network pharmacological-based investigation of the system-level mechanisms underlying the hematopoietic activity of Samul-tang, which is an herbal formula consisting of four herbal medicines, including Angelicae Gigantis Radix, Rehmanniae Radix Preparata, Paeoniae Radix Alba, and Cnidii Rhizoma. In silico analysis of the absorption-distribution-metabolism-excretion model identified 16 active phytochemical compounds contained in Samul-tang that may target 158 genes/proteins associated with myelosuppression to exert pharmacological effects. Functional enrichment analysis suggested that the targets of Samul-tang were significantly enriched in multiple pathways closely related to the hematopoiesis and myelosuppression development, including the PI3K-Akt, MAPK, IL-17, TNF, FoxO, HIF-1, NF-kappa B, and p53 signaling pathways. Our study provides novel evidence regarding the system-level mechanisms underlying the hematopoiesis-promoting effect of herbal medicines for hematological disorder treatment.
Collapse
|
5
|
Zhang K, Huang W, Li H, Xie M, Wang J. Ultrasensitive detection of hERG potassium channel in single-cell with photocleavable and entropy-driven reactions by using an electrochemical biosensor. Biosens Bioelectron 2019; 132:310-318. [DOI: 10.1016/j.bios.2019.02.065] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 02/26/2019] [Indexed: 12/23/2022]
|
6
|
Li X, Wilson AF, Du W, Pang Q. Cell-Cycle-Specific Function of p53 in Fanconi Anemia Hematopoietic Stem and Progenitor Cell Proliferation. Stem Cell Reports 2018; 10:339-346. [PMID: 29307578 PMCID: PMC5830889 DOI: 10.1016/j.stemcr.2017.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/18/2022] Open
Abstract
Overactive p53 has been proposed as an important pathophysiological factor for bone marrow failure syndromes, including Fanconi anemia (FA). Here, we report a p53-dependent effect on hematopoietic stem and progenitor cell (HSPC) proliferation in mice deficient for the FA gene Fanca. Deletion of p53 in Fanca-/- mice leads to replicative exhaustion of the hematopoietic stem cell (HSC) in transplant recipients. Using Fanca-/- HSCs expressing the separation-of-function mutant p53515C transgene, which selectively impairs the p53 function in apoptosis but keeps its cell-cycle checkpoint activities intact, we show that the p53 cell-cycle function is specifically required for the regulation of Fanca-/- HSC proliferation. Our results demonstrate that p53 plays a compensatory role in preventing FA HSCs from replicative exhaustion and suggest a cautious approach to manipulating p53 signaling as a therapeutic utility in FA.
Collapse
Affiliation(s)
- Xiaoli Li
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Andrew F Wilson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Wei Du
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA.
| | - Qishen Pang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
7
|
Kurre P. Hematopoietic development: a gap in our understanding of inherited bone marrow failure. Exp Hematol 2017; 59:1-8. [PMID: 29248612 DOI: 10.1016/j.exphem.2017.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/26/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022]
Abstract
Inherited bone marrow failure syndromes (IBMFS) represent a heterogeneous group of multisystem disorders that typically present with cytopenia in early childhood. Efforts to understand the underlying hematopoietic stem cell (HSC) losses have generally focused on postnatal hematopoiesis. However, reflecting the role of many of the involved genes in core cellular functions and the diverse nonhematologic abnormalities seen in patients at birth, studies have begun to explore IBMFS manifestations during fetal development. Here, I consider the current evidence for fetal deficits in the HSC pool and highlight emerging concepts regarding the origins and unique pathophysiology of hematopoietic failure in IBMFS.
Collapse
Affiliation(s)
- Peter Kurre
- Department of Pediatrics, Papé Family Pediatric Research Institute, Pediatric Blood & Cancer Biology Program, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
8
|
The combination of reduced MCL-1 and standard chemotherapeutics is tolerable in mice. Cell Death Differ 2017; 24:2032-2043. [PMID: 28800129 DOI: 10.1038/cdd.2017.125] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 06/14/2017] [Accepted: 07/03/2017] [Indexed: 02/06/2023] Open
Abstract
A common therapeutic strategy to combat human cancer is the use of combinations of drugs, each targeting different cellular processes or vulnerabilities. Recent studies suggest that addition of an MCL-1 inhibitor to such anticancer drug treatments could be an attractive therapeutic strategy. Thus, it is of great interest to understand whether combinations of conventional anticancer drugs with an MCL-1 inhibitor will be tolerable and efficacious. In order to mimic the combination of MCL-1 inhibition with other cancer therapeutics, we treated Mcl-1+/- heterozygous mice, which have a ~50% reduction in MCL-1 protein in their cells, with a broad range of chemotherapeutic drugs. Careful monitoring of treated mice revealed that a wide range of chemotherapeutic drugs had no significant effect on the general well-being of Mcl-1+/- mice with no overt damage to a broad range of tissues, including the haematopoietic compartment, heart, liver and kidney. These results indicate that MCL-1 inhibition may represent a tolerable strategy in cancer therapy, even when combined with select cytotoxic drugs.
Collapse
|
9
|
Muntión S, Ramos TL, Diez-Campelo M, Rosón B, Sánchez-Abarca LI, Misiewicz-Krzeminska I, Preciado S, Sarasquete ME, de las Rivas J, González M, Sánchez-Guijo F, del Cañizo MC. Microvesicles from Mesenchymal Stromal Cells Are Involved in HPC-Microenvironment Crosstalk in Myelodysplastic Patients. PLoS One 2016; 11:e0146722. [PMID: 26836120 PMCID: PMC4737489 DOI: 10.1371/journal.pone.0146722] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 12/20/2015] [Indexed: 01/28/2023] Open
Abstract
Exosomes/microvesicles (MVs) provide a mechanism of intercellular communication. Our hypothesis was that mesenchymal stromal cells (MSC) from myelodysplastic syndrome (MDS) patients could modify CD34+ cells properties by MVs. They were isolated from MSC from MDS patients and healthy donors (HD). MVs from 30 low-risk MDS patients and 27 HD were purified by ExoQuick-TC™ or ultracentrifugation and identified by transmission electron microscopy, flow cytometry (FC) and western blot for CD63. Incorporation of MVs into CD34+ cells was analyzed by FC, and confocal and fluorescence microscopy. Changes in hematopoietic progenitor cell (HPC) properties were assessed from modifications in microRNAs and gene expression in CD34+ cells as well as viability and clonogenic assays of CD34+ cells after MVs incorporation. Some microRNAs were overexpressed in MVs from patients MSC and two of them, miR-10a and miR-15a, were confirmed by RT-PCR. These microRNAs were transferred to CD34+ cells, modifying the expression of MDM2 and P53 genes, which was evaluated by RT-PCR and western blot. Finally, examining CD34+ cells properties after incorporation, higher cell viability (p = 0.025) and clonogenic capacity (p = 0.037) were observed when MVs from MDS patients were incorporated. In summary, we show that BM-MSC release MVs with a different cargo in MDS patients compared with HD. These structures are incorporated into HPC and modify their properties.
Collapse
Affiliation(s)
- Sandra Muntión
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
- Red Nacional de Terapia Celular (TerCel), Instituto Nacional de Salud Carlos III, Madrid, Spain
- IBSAL-Hospital Universitario Salamanca, Salamanca, Spain
| | - Teresa L. Ramos
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL-Hospital Universitario Salamanca, Salamanca, Spain
| | - María Diez-Campelo
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
- Red Nacional de Terapia Celular (TerCel), Instituto Nacional de Salud Carlos III, Madrid, Spain
- IBSAL-Hospital Universitario Salamanca, Salamanca, Spain
| | - Beatriz Rosón
- Red Nacional de Terapia Celular (TerCel), Instituto Nacional de Salud Carlos III, Madrid, Spain
| | - Luis Ignacio Sánchez-Abarca
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
- Red Nacional de Terapia Celular (TerCel), Instituto Nacional de Salud Carlos III, Madrid, Spain
- Centro de Investigación del Cáncer-IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
- IBSAL-Hospital Universitario Salamanca, Salamanca, Spain
| | - Irena Misiewicz-Krzeminska
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Centro de Investigación del Cáncer-IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
- National Medicines Institute, Warsaw, Poland
| | - Silvia Preciado
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
- Red Nacional de Terapia Celular (TerCel), Instituto Nacional de Salud Carlos III, Madrid, Spain
- Centro de Investigación del Cáncer-IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
- IBSAL-Hospital Universitario Salamanca, Salamanca, Spain
| | - María-Eugenia Sarasquete
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL-Hospital Universitario Salamanca, Salamanca, Spain
| | - Javier de las Rivas
- Red Nacional de Terapia Celular (TerCel), Instituto Nacional de Salud Carlos III, Madrid, Spain
| | - Marcos González
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Centro de Investigación del Cáncer-IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
- IBSAL-Hospital Universitario Salamanca, Salamanca, Spain
| | - Fermín Sánchez-Guijo
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
- Red Nacional de Terapia Celular (TerCel), Instituto Nacional de Salud Carlos III, Madrid, Spain
- Centro de Investigación del Cáncer-IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
- IBSAL-Hospital Universitario Salamanca, Salamanca, Spain
| | - María-Consuelo del Cañizo
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
- Red Nacional de Terapia Celular (TerCel), Instituto Nacional de Salud Carlos III, Madrid, Spain
- Centro de Investigación del Cáncer-IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
- IBSAL-Hospital Universitario Salamanca, Salamanca, Spain
- * E-mail:
| |
Collapse
|
10
|
Caspase-9 is required for normal hematopoietic development and protection from alkylator-induced DNA damage in mice. Blood 2014; 124:3887-95. [PMID: 25349173 DOI: 10.1182/blood-2014-06-582551] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Apoptosis and the DNA damage responses have been implicated in hematopoietic development and differentiation, as well as in the pathogenesis of myelodysplastic syndromes (MDS) and leukemia. However, the importance of late-stage mediators of apoptosis in hematopoiesis and leukemogenesis has not been elucidated. Here, we examine the role of caspase-9 (Casp9), the initiator caspase of the intrinsic apoptotic cascade, in murine fetal and adult hematopoiesis. Casp9 deficiency resulted in decreased erythroid and B-cell progenitor abundance and impaired function of hematopoietic stem cells after transplantation. Mouse bone marrow chimeras lacking Casp9 or its cofactor Apaf1 developed low white blood cell counts, decreased B-cell numbers, anemia, and reduced survival. Defects in apoptosis have also been previously implicated in susceptibility to therapy-related leukemia, a disease caused by exposure to DNA-damaging chemotherapy. We found that the burden of DNA damage was increased in Casp9-deficient cells after exposure to the alkylator, N-ethyl-nitrosourea (ENU). Furthermore, exome sequencing revealed that oligoclonal hematopoiesis emerged in Casp9-deficient bone marrow chimeras after alkylator exposure. Taken together, these findings suggest that defects in apoptosis could be a key step in the pathogenesis of alkylator-associated secondary malignancies.
Collapse
|
11
|
Belotte J, Fletcher NM, Alexis M, Morris RT, Munkarah AR, Diamond MP, Saed GM. Sox2 gene amplification significantly impacts overall survival in serous epithelial ovarian cancer. Reprod Sci 2014; 22:38-46. [PMID: 25038052 DOI: 10.1177/1933719114542021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epithelial ovarian cancer (EOC) is the deadliest gynecologic cancer. Recently, the existence of ovarian cancer stem cells has been reported. Sox2, Nanog and Oct4 are key markers of "stemness". The objective of this study was to determine whether Sox2, Nanog, and Oct4 are associated with EOC and poor outcome. The expression of these markers was assessed by immunofluorescence staining and real-time RT-PCR in human EOC cell lines MDAH-2774 and SKOV-3, while the cancer genome atlas (TCGA) dataset was analyzed for associations with survival. Sox2, Nanog and Oct4 (POU5F1) were all detected by immunofluorescence staining and these results were confirmed by real-time RT-PCR. The TCGA dataset revealed a 26%, 9%, and 6% amplification of Sox2, Nanog and POU5F1, respectively. Additionally, K-M survival analyses showed a significant median overall survival difference (41 versus 48.3 months, P = .01) for Sox2 amplification, but not for Nanog (44.1 versus 36.2 months, P > .05) and POU5F1 (43.5 versus 45.0 months, P > .05). Our results suggest that Sox2 gene amplification significantly influences overall survival.
Collapse
Affiliation(s)
- Jimmy Belotte
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nicole M Fletcher
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mitchell Alexis
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert T Morris
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Adnan R Munkarah
- Department of Obstetrics and Gynecology, Henry Ford Health System, Detroit, MI, USA
| | - Michael P Diamond
- Department of Obstetrics and Gynecology, Georgia Regents University, Augusta, GA, USA
| | - Ghassan M Saed
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
12
|
Siemens H, Jackstadt R, Kaller M, Hermeking H. Repression of c-Kit by p53 is mediated by miR-34 and is associated with reduced chemoresistance, migration and stemness. Oncotarget 2014; 4:1399-415. [PMID: 24009080 PMCID: PMC3824539 DOI: 10.18632/oncotarget.1202] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The c-Kit receptor tyrosine kinase is commonly over-expressed in different types of cancer. p53 activation is known to result in the down-regulation of c-Kit. However, the underlying mechanism has remained unknown. Here, we show that the p53-induced miR-34 microRNA family mediates repression of c-Kit by p53 via a conserved seed-matching sequence in the c-Kit 3'-UTR. Ectopic miR-34a resulted in a decrease in Erk signaling and transformation, which was dependent on the down-regulation of c-Kit expression. Furthermore, ectopic expression of c-Kit conferred resistance of colorectal cancer (CRC) cells to treatment with 5-fluorouracil (5-FU), whereas ectopic miR-34a sensitized the cells to 5-FU. After stimulation with c-Kit ligand/stem cell factor (SCF) Colo320 CRC cells displayed increased migration/invasion, whereas ectopic miR-34a inhibited SCF-induced migration/invasion. Activation of a conditional c-Kit allele induced several stemness markers in DLD-1 CRC cells. In primary CRC samples elevated c-Kit expression also showed a positive correlation with markers of stemness, such as Lgr5, CD44, OLFM4, BMI-1 and β-catenin. On the contrary, activation of a conditional miR-34a allele in DLD-1 cells diminished the expression of c-Kit and several stemness markers (CD44, Lgr5 and BMI-1) and suppressed sphere formation. MiR-34a also suppressed enhanced sphere-formation after exposure to SCF. Taken together, our data establish c-Kit as a new direct target of miR-34 and demonstrate that this regulation interferes with several c-Kit-mediated effects on cancer cells. Therefore, this regulation may be potentially relevant for future diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Helge Siemens
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University München, D-80337 Munich, Germany
| | | | | | | |
Collapse
|
13
|
Piccoli C, Agriesti F, Scrima R, Falzetti F, Di Ianni M, Capitanio N. To breathe or not to breathe: the haematopoietic stem/progenitor cells dilemma. Br J Pharmacol 2014; 169:1652-71. [PMID: 23714011 DOI: 10.1111/bph.12253] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 05/11/2013] [Accepted: 05/16/2013] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Adult haematopoietic stem/progenitor cells (HSPCs) constitute the lifespan reserve for the generation of all the cellular lineages in the blood. Although massive progress in identifying the cluster of master genes controlling self-renewal and multipotency has been achieved in the past decade, some aspects of the physiology of HSPCs still need to be clarified. In particular, there is growing interest in the metabolic profile of HSPCs in view of their emerging role as determinants of cell fate. Indeed, stem cells and progenitors have distinct metabolic profiles, and the transition from stem to progenitor cell corresponds to a critical metabolic change, from glycolysis to oxidative phosphorylation. In this review, we summarize evidence, reported in the literature and provided by our group, highlighting the peculiar ability of HSPCs to adapt their mitochondrial oxidative/bioenergetic metabolism to survive in the hypoxic microenvironment of the endoblastic niche and to exploit redox signalling in controlling the balance between quiescence versus active cycling and differentiation. Especial prominence is given to the interplay between hypoxia inducible factor-1, globins and NADPH oxidases in managing the mitochondrial dioxygen-related metabolism and biogenesis in HSPCs under different ambient conditions. A mechanistic model is proposed whereby 'mitochondrial differentiation' is a prerequisite in uncommitted stem cells, paving the way for growth/differentiation factor-dependent processes. Advancing the understanding of stem cell metabolism will, hopefully, help to (i) improve efforts to maintain, expand and manipulate HSPCs ex vivo and realize their potential therapeutic benefits in regenerative medicine; (ii) reprogramme somatic cells to generate stem cells; and (iii) eliminate, selectively, malignant stem cells. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
Affiliation(s)
- C Piccoli
- Department of Medical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | | | | | | | | | | |
Collapse
|
14
|
Ellis SR. Nucleolar stress in Diamond Blackfan anemia pathophysiology. Biochim Biophys Acta Mol Basis Dis 2014; 1842:765-8. [PMID: 24412987 DOI: 10.1016/j.bbadis.2013.12.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 12/23/2013] [Accepted: 12/31/2013] [Indexed: 01/05/2023]
Abstract
Diamond Blackfan anemia is a red cell hypoplasia that typically presents within the first year of life. Most cases of Diamond Blackfan anemia are caused by ribosome assembly defects linked to haploinsufficiency for structural proteins of either ribosomal subunit. Nucleolar stress associated with abortive ribosome assembly leads to p53 activation via the interaction of free ribosomal proteins with HDM2, a negative regulator of p53. Significant challenges remain in linking this nucleolar stress signaling pathway to the clinical features of Diamond Blackfan anemia. Defining aspects of disease presentation may relate to developmental and physiological triggers that work in conjunction with nucleolar stress signaling to heighten the p53 response in the developing erythron after birth. The growing number of ribosomopathies provides additional challenges for linking molecular mechanisms with clinical phenotypes. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.
Collapse
Affiliation(s)
- Steven R Ellis
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
15
|
Basova P, Pospisil V, Savvulidi F, Burda P, Vargova K, Stanek L, Dluhosova M, Kuzmova E, Jonasova A, Steidl U, Laslo P, Stopka T. Aggressive acute myeloid leukemia in PU.1/p53 double-mutant mice. Oncogene 2013; 33:4735-45. [PMID: 24121269 DOI: 10.1038/onc.2013.414] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 08/23/2013] [Accepted: 09/02/2013] [Indexed: 01/02/2023]
Abstract
PU.1 downregulation within hematopoietic stem and progenitor cells (HSPCs) is the primary mechanism for the development of acute myeloid leukemia (AML) in mice with homozygous deletion of the upstream regulatory element (URE) of PU.1 gene. p53 is a well-known tumor suppressor that is often mutated in human hematologic malignancies including AML and adds to their aggressiveness; however, its genetic deletion does not cause AML in mouse. Deletion of p53 in the PU.1(ure/ure) mice (PU.1(ure/ure)p53(-/-)) results in more aggressive AML with shortened overall survival. PU.1(ure/ure)p53(-/-) progenitors express significantly lower PU.1 levels. In addition to URE deletion we searched for other mechanisms that in the absence of p53 contribute to decreased PU.1 levels in PU.1(ure/ure)p53(-/-) mice. We found involvement of Myb and miR-155 in downregulation of PU.1 in aggressive murine AML. Upon inhibition of either Myb or miR-155 in vitro the AML progenitors restore PU.1 levels and lose leukemic cell growth similarly to PU.1 rescue. The MYB/miR-155/PU.1 axis is a target of p53 and is activated early after p53 loss as indicated by transient p53 knockdown. Furthermore, deregulation of both MYB and miR-155 coupled with PU.1 downregulation was observed in human AML, suggesting that MYB/miR-155/PU.1 mechanism may be involved in the pathogenesis of AML and its aggressiveness characterized by p53 mutation.
Collapse
Affiliation(s)
- P Basova
- 1] Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic [2] Department of Experimental Biomodels, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - V Pospisil
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - F Savvulidi
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - P Burda
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - K Vargova
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - L Stanek
- 1] Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic [2] Department of Pathology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - M Dluhosova
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - E Kuzmova
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - A Jonasova
- 1] Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic [2] Department of Medicine-Haematology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - U Steidl
- Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| | - P Laslo
- Section of Experimental Haematology, Leeds Institute of Cancer and Pathology, St James's University Hospital, University of Leeds, Leeds, UK
| | - T Stopka
- 1] Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic [2] Department of Medicine-Haematology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
16
|
Romeo F, Costanzo F, Agostini M. Embryonic stem cells and inducible pluripotent stem cells: two faces of the same coin? Aging (Albany NY) 2013; 4:878-86. [PMID: 23248145 PMCID: PMC3615155 DOI: 10.18632/aging.100513] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Embryonic stem cells (ESCs) are derived from the inner cell mass of the blastocysts and are characterized by the ability to renew themselves (self-renewal) and the capability to generate all the cells within the human body. In contrast, inducible pluripotent stem cells (iPSCs) are generated by transfection of four transcription factors in somatic cells. Like embryonic stem cells, they are able to self-renew and differentiate. Because of these features, both ESCs and iPSCs, are under intense clinical investigation for cell-based therapy. In this review, we revisit stem cell biology and add a new layer of complexity. In particular, we will highlight some of the complexities of the system, but also where there may be therapeutic potential for modulation of intrinsic stem cells and where particular caution may be needed in terms of cell transplantation therapies.
Collapse
Affiliation(s)
- Francesco Romeo
- Department of Experimental and Clinical Medicine, Magna Gracia University of Catanzaro, Salvatore Venuta Campus, 88100 Catanzaro, Italy
| | | | | |
Collapse
|
17
|
Yamada T, Park CS, Lacorazza HD. Genetic control of quiescence in hematopoietic stem cells. Cell Cycle 2013; 12:2376-83. [PMID: 23839041 PMCID: PMC3841317 DOI: 10.4161/cc.25416] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 06/13/2013] [Accepted: 06/14/2013] [Indexed: 01/08/2023] Open
Abstract
Cellular quiescence is a reversible cell cycle arrest that is poised to re-enter the cell cycle in response to a combination of cell-intrinsic factors and environmental cues. In hematopoietic stem cells, a coordinated balance between quiescence and differentiating proliferation ensures longevity and prevents both genetic damage and stem cell exhaustion. However, little is known about how all these processes are integrated at the molecular level. We will briefly review the environmental and intrinsic control of stem cell quiescence and discuss a new model that involves a protein-to-protein interaction between G0S2 and the phospho-nucleoprotein nucleolin in the cytosol.
Collapse
Affiliation(s)
- Takeshi Yamada
- Department of Pathology & Immunology; Baylor College of Medicine; Texas Children’s Hospital; Houston, TX USA
| | - Chun Shik Park
- Department of Pathology & Immunology; Baylor College of Medicine; Texas Children’s Hospital; Houston, TX USA
| | - H Daniel Lacorazza
- Department of Pathology & Immunology; Baylor College of Medicine; Texas Children’s Hospital; Houston, TX USA
| |
Collapse
|
18
|
Haupt S, Mitchell C, Corneille V, Shortt J, Fox S, Pandolfi PP, Castillo-Martin M, Bonal DM, Cordon-Cardo C, Lozano G, Haupt Y. Loss of PML cooperates with mutant p53 to drive more aggressive cancers in a gender-dependent manner. Cell Cycle 2013; 12:1722-31. [PMID: 23656786 DOI: 10.4161/cc.24805] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED p53 mutations and downregulation of promyelocytic leukemia (PML) are common genetic alterations in human cancers. In healthy cells these two key tumor suppressors exist in a positive regulatory loop, promoting cell death and cellular senescence. However, the influence of their interplay on tumorigenesis has not been explored directly in vivo. The contribution of PML to mutant p53 driven cancer was evaluated in a mouse model harboring a p53 mutation (p53 (wild-type/R172H) ) that recapitulates a frequent p53 mutation (p53 (R175H) ) in human sporadic and Li-Fraumeni cancers. These mice with PML displayed perturbation of the hematopoietic compartment, manifested either as lymphoma or extramedullary hematopoiesis (EMH). EMH was associated with peripheral blood leucocytosis and macrocytic anemia, suggestive of myeloproliferative- myelodysplastic overlap. In contrast, a complete loss of PML from these mice resulted in a marked alteration in tumor profile. While the incidence of lymphomas was unaltered, EMH was not detected and the majority of mice succumbed to sarcomas. Further, males lacking PML exhibited a high incidence of soft tissue sarcomas and reduced survival, while females largely developed osteosarcomas, without impact on survival. Together, these findings demonstrate that PML is an important tumor suppressor dictating disease development in a pertinent mouse model of human cancer. KEY POINTS (1) A mutant p53 allele disrupts hematopoiesis in mice, by promoting lymphomas and myeloproliferative / myelodysplastic overlap. (2) Coincidental p53 allele mutation and PML loss shifts the tumor profile toward sarcoma formation, which is paralleled in human leiomyosarcomas (indicated by immunohistochemistry; IHC).
Collapse
Affiliation(s)
- Sue Haupt
- Research Division, Peter MacCallum Cancer Centre, East Melbourne, VIC Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Wang K, Zhang T, Dong Q, Nice EC, Huang C, Wei Y. Redox homeostasis: the linchpin in stem cell self-renewal and differentiation. Cell Death Dis 2013; 4:e537. [PMID: 23492768 PMCID: PMC3613828 DOI: 10.1038/cddis.2013.50] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Stem cells are characterized by their unique ability of self-renewal to maintain the so-called stem cell pool. Over the past decades, reactive oxygen species (ROS) have been recognized as toxic aerobic metabolism byproducts that are harmful to stem cells, leading to DNA damage, senescence or cell death. Recently, a growing body of literature has shown that stem cells reside in redox niches with low ROS levels. The balance of Redox homeostasis facilitates stem cell self-renewal by an intricate network. Thus, to fully decipher the underlying molecular mechanisms involved in the maintenance of stem cell self-renewal, it is critical to address the important role of redox homeostasis in the regulation of self-renewal and differentiation of stem cells. In this regard, we will discuss the regulatory mechanisms involved in the subtly orchestrated balance of redox status in stem cells by scavenger antioxidant enzyme systems that are well monitored by the hypoxia niches and crucial redox regulators including forkhead homeobox type O family (FoxOs), apurinic/apyrimidinic (AP) endonuclease1/redox factor-1 (APE1/Ref-1), nuclear factor erythroid-2-related factor 2 (Nrf2) and ataxia telangiectasia mutated (ATM). We will also introduce several pivotal ROS-sensitive molecules, such as hypoxia-inducible factors, p38 mitogen-activated protein kinase (p38) and p53, involved in the redox-regulated stem cell self-renewal. Specifically, all the aforementioned molecules can act as ‘redox sensors' by virtue of redox modifications of their cysteine residues, which are critically important in the control of protein function. Given the importance of redox homeostasis in the regulation of stem cell self-renewal, understanding the underlying molecular mechanisms involved will provide important new insights into stem cell biology.
Collapse
Affiliation(s)
- Kui Wang
- The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | | | | | | | | | | |
Collapse
|
20
|
Jackson JG, Lozano G. The mutant p53 mouse as a pre-clinical model. Oncogene 2013; 32:4325-30. [PMID: 23318424 DOI: 10.1038/onc.2012.610] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/24/2012] [Accepted: 11/07/2012] [Indexed: 11/09/2022]
Abstract
The p53 tumor-suppressor pathway is dismantled in the development of most cancers. Mice with various p53 mutant alleles either singly or in combination with other genetic alterations are predisposed to tumor development. Here, we review studies utilizing p53 mutant mice that have recapitulated and informed clinical observations. These studies have demonstrated the p53 contribution, sometimes beneficial and sometimes detrimental, to treatment response in lymphomas, and lung and breast cancers. Further, we examine how p53 mutant mouse models have been used to test the efficacy of p53 reactivation as a therapeutic strategy.
Collapse
Affiliation(s)
- J G Jackson
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
21
|
Urao N, Ushio-Fukai M. Redox regulation of stem/progenitor cells and bone marrow niche. Free Radic Biol Med 2013; 54:26-39. [PMID: 23085514 PMCID: PMC3637653 DOI: 10.1016/j.freeradbiomed.2012.10.532] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 10/02/2012] [Accepted: 10/05/2012] [Indexed: 01/19/2023]
Abstract
Bone marrow (BM)-derived stem and progenitor cell functions including self-renewal, differentiation, survival, migration, proliferation, and mobilization are regulated by unique cell-intrinsic and -extrinsic signals provided by their microenvironment, also termed the "niche." Reactive oxygen species (ROS), especially hydrogen peroxide (H(2)O(2)), play important roles in regulating stem and progenitor cell functions in various physiologic and pathologic responses. The low level of H(2)O(2) in quiescent hematopoietic stem cells (HSCs) contributes to maintaining their "stemness," whereas a higher level of H(2)O(2) within HSCs or their niche promotes differentiation, proliferation, migration, and survival of HSCs or stem/progenitor cells. Major sources of ROS are NADPH oxidase and mitochondria. In response to ischemic injury, ROS derived from NADPH oxidase are increased in the BM microenvironment, which is required for hypoxia and hypoxia-inducible factor-1α expression and expansion throughout the BM. This, in turn, promotes progenitor cell expansion and mobilization from BM, leading to reparative neovascularization and tissue repair. In pathophysiological states such as aging, atherosclerosis, heart failure, hypertension, and diabetes, excess amounts of ROS create an inflammatory and oxidative microenvironment, which induces cell damage and apoptosis of stem and progenitor cells. Understanding the molecular mechanisms of how ROS regulate the functions of stem and progenitor cells and their niche in physiological and pathological conditions will lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|
22
|
Agasti SS, Liong M, Peterson VM, Lee H, Weissleder R. Photocleavable DNA barcode-antibody conjugates allow sensitive and multiplexed protein analysis in single cells. J Am Chem Soc 2012; 134:18499-502. [PMID: 23092113 DOI: 10.1021/ja307689w] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
DNA barcoding is an attractive technology, as it allows sensitive and multiplexed target analysis. However, DNA barcoding of cellular proteins remains challenging, primarily because barcode amplification and readout techniques are often incompatible with the cellular microenvironment. Here we describe the development and validation of a photocleavable DNA barcode-antibody conjugate method for rapid, quantitative, and multiplexed detection of proteins in single live cells. Following target binding, this method allows DNA barcodes to be photoreleased in solution, enabling easy isolation, amplification, and readout. As a proof of principle, we demonstrate sensitive and multiplexed detection of protein biomarkers in a variety of cancer cells.
Collapse
Affiliation(s)
- Sarit S Agasti
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical, Boston, 02114, United States
| | | | | | | | | |
Collapse
|
23
|
Vicente-Dueñas C, González-Herrero I, García Cenador MB, García Criado FJ, Sánchez-García I. Loss of p53 exacerbates multiple myeloma phenotype by facilitating the reprogramming of hematopoietic stem/progenitor cells to malignant plasma cells by MafB. Cell Cycle 2012; 11:3896-900. [PMID: 22983007 DOI: 10.4161/cc.22186] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is a serious, mostly incurable human cancer of malignant plasma cells. Chromosomal translocations affecting MAFB are present in a significant percentage of multiple myeloma patients. Genetically engineered Sca1-MafB mice, in which MafB expression is limited to hematopoietic stem/progenitor cells (HS/P-Cs), display the phenotypic features of MM. Contrary to many other types of cancer, it is not yet known if the p53 gene plays any essential role in the pathogenesis of this disease. Here, we show, taking advantage of the Sca1-MafB MM mouse model, that loss of p53 does not rescue the multiple myeloma disease, but instead accelerates its development and exacerbates the MM phenotype. Therefore, the efficiency of the MafB-induced MM reprogramming of normal HS/P-Cs to terminally differentiated malignant plasma cells is enhanced by p53 deficiency, in analogy to what happens in reprogramming to pluripotency. These results raise caution about interfering with p53 function when treating multiple myeloma.
Collapse
Affiliation(s)
- Carolina Vicente-Dueñas
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain.
| | | | | | | | | |
Collapse
|