51
|
Tanno B, Leonardi S, Babini G, Giardullo P, De Stefano I, Pasquali E, Saran A, Mancuso M. Nanog-driven cell-reprogramming and self-renewal maintenance in Ptch1 +/- granule cell precursors after radiation injury. Sci Rep 2017; 7:14238. [PMID: 29079783 PMCID: PMC5660207 DOI: 10.1038/s41598-017-14506-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/11/2017] [Indexed: 12/31/2022] Open
Abstract
Medulloblastoma (MB) is the most common pediatric brain tumor, comprising four distinct molecular variants, one of which characterized by activation of the Sonic Hedgehog (SHH) pathway, driving 25–30% of sporadic MB. SHH-dependent MBs arise from granule cell precursors (GCPs), are fatal in 40–70% of cases and radioresistance strongly contributes to poor prognosis and tumor recurrence. Patched1 heterozygous (Ptch1+/−) mice, carrying a germ-line heterozygous inactivating mutation in the Ptch1 gene, the Shh receptor and negative regulator of the pathway, are uniquely susceptible to MB development after radiation damage in neonatal cerebellum. Here, we irradiated ex-vivo GCPs isolated from cerebella of neonatal WT and Ptch1+/− mice. Our results highlight a less differentiated status of Ptch1-mutated cells after irradiation, influencing DNA damage response. Increased expression levels of pluripotency genes Nanog, Oct4 and Sal4, together with greater clonogenic potential, clearly suggest that radiation induces expansion of the stem-like cell compartment through cell-reprogramming and self-renewal maintenance, and that this mechanism is strongly dependent on Nanog. These results contribute to clarify the molecular mechanisms that control radiation-induced Shh-mediated tumorigenesis and may suggest Nanog as a potential target to inhibit for adjuvant radiotherapy in treatment of SHH-dependent MB.
Collapse
Affiliation(s)
- Barbara Tanno
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | - Simona Leonardi
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | | | - Paola Giardullo
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy.,Department of Sciences, Roma Tre University, Rome, Italy
| | - Ilaria De Stefano
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy
| | - Emanuela Pasquali
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | - Anna Saran
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy.
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy.
| |
Collapse
|
52
|
Huang WY, Lai SF, Chiu HY, Chang M, Plikus MV, Chan CC, Chen YT, Tsao PN, Yang TL, Lee HS, Chi P, Lin SJ. Mobilizing Transit-Amplifying Cell-Derived Ectopic Progenitors Prevents Hair Loss from Chemotherapy or Radiation Therapy. Cancer Res 2017; 77:6083-6096. [PMID: 28939680 DOI: 10.1158/0008-5472.can-17-0667] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 08/02/2017] [Accepted: 09/14/2017] [Indexed: 12/15/2022]
Abstract
Genotoxicity-induced hair loss from chemotherapy and radiotherapy is often encountered in cancer treatment, and there is a lack of effective treatment. In growing hair follicles (HF), quiescent stem cells (SC) are maintained in the bulge region, and hair bulbs at the base contain rapidly dividing, yet genotoxicity-sensitive transit-amplifying cells (TAC) that maintain hair growth. How genotoxicity-induced HF injury is repaired remains unclear. We report here that HFs mobilize ectopic progenitors from distinct TAC compartments for regeneration in adaptation to the severity of dystrophy induced by ionizing radiation (IR). Specifically, after low-dose IR, keratin 5+ basal hair bulb progenitors, rather than bulge SCs, were quickly activated to replenish matrix cells and regenerated all concentric layers of HFs, demonstrating their plasticity. After high-dose IR, when both matrix and hair bulb cells were depleted, the surviving outer root sheath cells rapidly acquired an SC-like state and fueled HF regeneration. Their progeny then homed back to SC niche and supported new cycles of HF growth. We also revealed that IR induced HF dystrophy and hair loss and suppressed WNT signaling in a p53- and dose-dependent manner. Augmenting WNT signaling attenuated the suppressive effect of p53 and enhanced ectopic progenitor proliferation after genotoxic injury, thereby preventing both IR- and cyclophosphamide-induced alopecia. Hence, targeted activation of TAC-derived progenitor cells, rather than quiescent bulge SCs, for anagen HF repair can be a potential approach to prevent hair loss from chemotherapy and radiotherapy. Cancer Res; 77(22); 6083-96. ©2017 AACR.
Collapse
Affiliation(s)
- Wen-Yen Huang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Shih-Fan Lai
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.,Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Hsien-Yi Chiu
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.,Department of Dermatology, Hsin-Chu Branch, National Taiwan University Hospital, Hsin-Chu City, Taiwan.,Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Michael Chang
- Sophie Davis School of Biomedical Education, City University of New York, New York, New York
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center and Center for Complex Biological Systems, University of California, Irvine, Irvine, California
| | - Chih-Chieh Chan
- Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - You-Tzung Chen
- Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Po-Nien Tsao
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsung-Lin Yang
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsuan-Shu Lee
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.,Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Peter Chi
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Sung-Jan Lin
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan. .,Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
53
|
DNA damage in protective and adverse inflammatory responses: Friend of foe? Mech Ageing Dev 2017; 165:47-53. [DOI: 10.1016/j.mad.2016.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/13/2016] [Indexed: 11/17/2022]
|
54
|
Dall G, Risbridger G, Britt K. Mammary stem cells and parity-induced breast cancer protection- new insights. J Steroid Biochem Mol Biol 2017; 170:54-60. [PMID: 26907964 DOI: 10.1016/j.jsbmb.2016.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/09/2016] [Accepted: 02/18/2016] [Indexed: 11/26/2022]
Abstract
Parity (childbearing) significantly decreases a woman's risk of breast cancer and the protective effect is greater if the woman is younger and has more children. The mechanism/s of parity-induced protection are not known. Although several factors are postulated to play a role, we discuss how a reduction in the number of mammary stem cells (MaSCs) may lead to a reduction in breast cancer risk in parous women. Firstly we review the epidemiology linking childbearing to reduced breast cancer risk and discuss how additional births, a young age at first full term birth, and breastfeeding impact the protection. We then detail the mouse and human studies implicating MaSC in parity induced protection and the in-vivo work being performed in mice to directly investigate the effect of parity on MaSC. Finally we discuss the transplant and lineage tracing experiments assessing MaSC activity according to parity and the need to define if MaSC are indeed more carcinogen sensitive than mature mammary epithelial cells. Continuing and future studies attempting to define the parity induced mechanisms will aid in the development of preventative therapies.
Collapse
Affiliation(s)
- Genevieve Dall
- Metastasis Research Laboratory, Peter MacCallum Cancer Centre, 7 St Andrews Place, East Melbourne 3002, Australia; Department of Anatomy and Developmental Biology, Monash University Clayton, Wellington Rd 3800, Australia
| | - Gail Risbridger
- Department of Anatomy and Developmental Biology, Monash University Clayton, Wellington Rd 3800, Australia
| | - Kara Britt
- Metastasis Research Laboratory, Peter MacCallum Cancer Centre, 7 St Andrews Place, East Melbourne 3002, Australia; Department of Anatomy and Developmental Biology, Monash University Clayton, Wellington Rd 3800, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia.
| |
Collapse
|
55
|
A coordinated DNA damage response promotes adult quiescent neural stem cell activation. PLoS Biol 2017; 15:e2001264. [PMID: 28489848 PMCID: PMC5424956 DOI: 10.1371/journal.pbio.2001264] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/31/2017] [Indexed: 11/24/2022] Open
Abstract
Stem and differentiated cells frequently differ in their response to DNA damage, which can determine tissue sensitivity. By exploiting insight into the spatial arrangement of subdomains within the adult neural subventricular zone (SVZ) in vivo, we show distinct responses to ionising radiation (IR) between neural stem and progenitor cells. Further, we reveal different DNA damage responses between neonatal and adult neural stem cells (NSCs). Neural progenitors (transit amplifying cells and neuroblasts) but not NSCs (quiescent and activated) undergo apoptosis after 2 Gy IR. This response is cell type- rather than proliferation-dependent and does not appear to be driven by distinctions in DNA damage induction or repair capacity. Moreover, exposure to 2 Gy IR promotes proliferation arrest and differentiation in the adult SVZ. These 3 responses are ataxia telangiectasia mutated (ATM)-dependent and promote quiescent NSC (qNSC) activation, which does not occur in the subdomains that lack progenitors. Neuroblasts arising post-IR derive from activated qNSCs rather than irradiated progenitors, minimising damage compounded by replication or mitosis. We propose that rather than conferring sensitive cell death, apoptosis is a form of rapid cell death that serves to remove damaged progenitors and promote qNSC activation. Significantly, analysis of the neonatal (P5) SVZ reveals that although progenitors remain sensitive to apoptosis, they fail to efficiently arrest proliferation. Consequently, their repopulation occurs rapidly from irradiated progenitors rather than via qNSC activation. The response of stem cells to DNA damage is poorly understood, although there is increasing evidence that they respond distinctly to differentiated cells. We have monitored the different responses of adult neural stem and progenitor cells to exposure to X-ray irradiation. We see that progenitor cells activate apoptosis, undergo rapid proliferation arrest, and premature differentiation. However, quiescent stem cells do not activate radiation-induced apoptosis. Instead the responses of the progenitor cells promote the activation of these quiescent neural stem cells, thereby replenishing the neuroblasts. These responses and quiescent stem cell activation are dependent on the ataxia telangiectasia mutated (ATM) kinase. We propose that this coordinated response functions to remove damaged progenitor cells and to aid repopulation.
Collapse
|
56
|
Vitale I, Manic G, De Maria R, Kroemer G, Galluzzi L. DNA Damage in Stem Cells. Mol Cell 2017; 66:306-319. [DOI: 10.1016/j.molcel.2017.04.006] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/23/2017] [Accepted: 04/05/2017] [Indexed: 01/03/2023]
|
57
|
Biechonski S, Yassin M, Milyavsky M. DNA-damage response in hematopoietic stem cells: an evolutionary trade-off between blood regeneration and leukemia suppression. Carcinogenesis 2017; 38:367-377. [PMID: 28334174 DOI: 10.1093/carcin/bgx002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/11/2017] [Indexed: 12/12/2022] Open
Abstract
Self-renewing and multipotent hematopoietic stem cells (HSCs) maintain lifelong hematopoiesis. Their enormous regenerative potential coupled with lifetime persistence in the body, in contrast with the Progenitors, demand tight control of HSCs genome stability. Indeed, failure to accurately repair DNA damage in HSCs is associated with bone marrow failure and accelerated leukemogenesis. Recent observations exposed remarkable differences in several DNA-damage response (DDR) aspects between HSCs and Progenitors, especially in their DNA-repair capacities and susceptibility to apoptosis. Human HSCs in comparison with Progenitors exhibit delayed DNA double-strand break rejoining, persistent DDR signaling activation, higher sensitivity to the cytotoxic effects of ionizing radiation and attenuated expression of DNA-repair genes. Importantly, the distinct DDR of HSCs was also documented in mouse models. Nevertheless, physiological significance and the molecular basis of the HSCs-specific DDR features are only partially understood. Taking radiation-induced DDR as a paradigm, this review will focus on the current advances in understanding the role of cell-intrinsic DDR regulators and the cellular microenvironment in balancing stemness with genome stability. Pre-leukemia HSCs and clonal hematopoiesis evolvement will be discussed as an evolutionary compromise between the need for lifelong blood regeneration and DDR. Uniquely for this review, we outline the differences in HSCs-related DDR as highlighted by various experimental systems and attempt to provide their critical analysis.
Collapse
Affiliation(s)
- Shahar Biechonski
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Muhammad Yassin
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Michael Milyavsky
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
58
|
Cai J, Cai Y, Ma Q, Chang F, Xu L, Zhang G, Guo X. Association of p53 codon 72 polymorphism with susceptibility to hepatocellular carcinoma in a Chinese population from northeast Sichuan. Biomed Rep 2017; 6:217-222. [PMID: 28357076 DOI: 10.3892/br.2017.840] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/07/2016] [Indexed: 11/05/2022] Open
Abstract
The p53 tumor suppressor gene is key in tumor development and progression, and the single nucleotide polymorphism (SNP) of the p53 gene codon 72 (p53Arg/Pro) changes the structure of the protein. In addition, it affects its activity, which may affect cancer risk. The aim of the present study was to investigate the association between p53 codon 72 polymorphism and susceptibility to hepatocellular carcinoma (HCC) in a Chinese population from northeast Sichuan. A total of 342 HCC patients and 347 non-cancer control subjects were recruited, and the polymorphism of p53 codon 72 was measured by TaqMan® minor groove binder fluorescent quantitative polymerase chain reaction assay. The distribution frequency of p53 sites of arginine (Arg)/Arg, Arg/proline (Pro), Pro/Pro were 18.4, 48.8 and 32.8% in the control group, as compared with 18.7, 49.9 and 31.4% in the case group, which indicated that there was no difference between two groups (χ2=0.14; P=0.93). Upon further stratification with smoking, alcohol consumption, gender and hepatitis B virus (HBV) infection, no risk increasing genotype was identified. However, interactions between p53 codon 72 SNP and smoking, alcohol consumption and HBV infection may increase the risk of HCC [smoking odds ratio (OR), 2.00; 95% confidence interval (CI), 1.21-3.29; alcohol consumption OR, 1.87; 95% CI, 1.08-3.26; HBV infection OR, 1.84; 95% CI, 1.10-3.08]. No significant association was identified between p53 codon 72 polymorphism and HCC, and it may not have an independent effect on the susceptibility to HCC in a Chinese population from northeast Sichuan. However, interaction between genetic factors and environment exposure significantly increased the risk of HCC.
Collapse
Affiliation(s)
- Jiajing Cai
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China; Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yan Cai
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China; Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Qiang Ma
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China; Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Fan Chang
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Lei Xu
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guoyuan Zhang
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China; Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xiaolan Guo
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China; Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
59
|
Genetic polymorphisms and plasma levels of BCL11A contribute to the development of laryngeal squamous cell carcinoma. PLoS One 2017; 12:e0171116. [PMID: 28225775 PMCID: PMC5321498 DOI: 10.1371/journal.pone.0171116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 12/31/2016] [Indexed: 11/19/2022] Open
Abstract
Objective We investigated the association between B-cell lymphoma/leukaemia 11A (BCL11A) rs11886868 and rs4671393 polymorphism, plasma BCL11A concentration, and the hazard of developing laryngeal squamous cell carcinoma (LSCC). Participants and method In this research, 330 LSCC patients, 310 healthy controls, and 155 vocal leukoplakia patients were genotyped for the BCL11A (rs11886868 C/T and rs4671393 A/G) genotypes by pyrosequencing; the BCL11A concentration was measured using ELISA. Results LSCC Patients had a notably higher occurrence of CT at rs11886868 (OR = 2.64, P = 0.025) than the control group; they also had higher GG at rs4671393 (OR = 2.53, P = 0.018). Advanced (III and IV) stage LSCC patients had a notably greater frequency of CT at rs11886868 than those with initial (I and II) stage LSCC (OR = 2.71, P = 0.044 vs. OR = 2.58, P = 0.051). Additionally, there was a 1.59 fold increase in susceptibility for initial stage LSCC related to the G allele (AG/GG) at rs4671393 (P = 0.005); while for patients of advanced stage LSCC the OR was 1.73 (P = 0.002). Moreover, the OR of lymph node metastasis patients at rs4671393 G alleles was 2.41 (P < 0.01); it was 1.38 (P = 0.035) in patients without lymph metastasis. Patients with high incidences of the rs4671393 variation genotype had high plasma BCL11A levels. Conclusions BCL11A rs11886868 and rs4671393 genotype variations and correspondingly high BCL11A plasma levels are related to LSCC, besides, differences in plasma levels and genotype distribution may be related to lymph node metastasis status and the stage of LSCC.
Collapse
|
60
|
Schäfer P, Karl MO. Prospective purification and characterization of Müller glia in the mouse retina regeneration assay. Glia 2017; 65:828-847. [PMID: 28220544 DOI: 10.1002/glia.23130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 01/06/2023]
Abstract
Reactive gliosis is an umbrella term for various glia functions in neurodegenerative diseases and upon injury. Specifically, Müller glia (MG) in some species readily regenerate retinal neurons to restore vision loss after insult, whereas mammalian MG respond by reactive gliosis-a heterogeneous response which frequently includes cell hypertrophy and proliferation. Limited regeneration has been stimulated in mammals, with a higher propensity in young MG, and in vitro compared to in vivo, but the underlying processes are unknown. To facilitate studies on the mechanisms regulating and limiting glia functions, we developed a strategy to purify glia and their progeny by fluorescence-activated cell sorting. Dual-transgenic nuclear reporter mice, which label neurons and glia with red and green fluorescent proteins, respectively, have enabled MG enrichment up to 93% purity. We applied this approach to MG in a mouse retina regeneration ex vivo assay. Combined cell size and cell cycle analysis indicates that most MG hypertrophy and a subpopulation proliferates which, over time, become even larger in cell size than the ones that do not proliferate. MG undergo timed differential genomic changes in genes controlling stemness and neurogenic competence; and glial markers are downregulated. Genes that are potentially required for, or associated with, regeneration and reactive gliosis are differentially regulated by retina explant culture time, epidermal growth factor stimulation, and animal age. Thus, MG enrichment facilitates cellular and molecular studies which, in combination with the mouse retina regeneration assay, provide an experimental approach for deciphering mechanisms that possibly regulate reactive gliosis and limit regeneration in mammals.
Collapse
Affiliation(s)
- Patrick Schäfer
- TU Dresden, Center for Regenerative Therapies Dresden (CRTD), Fetscherstr. 107, Dresden, 01307, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Arnoldstr. 13, Dresden, 01307, Germany
| | - Mike O Karl
- TU Dresden, Center for Regenerative Therapies Dresden (CRTD), Fetscherstr. 107, Dresden, 01307, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Arnoldstr. 13, Dresden, 01307, Germany
| |
Collapse
|
61
|
Resistance for Genotoxic Damage in Mesenchymal Stromal Cells Is Increased by Hypoxia but Not Generally Dependent on p53-Regulated Cell Cycle Arrest. PLoS One 2017; 12:e0169921. [PMID: 28081228 PMCID: PMC5231334 DOI: 10.1371/journal.pone.0169921] [Citation(s) in RCA: 249] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/23/2016] [Indexed: 11/19/2022] Open
Abstract
Adult stem cells including multipotent mesenchymal stromal cells (MSC) acquire a high amount of DNA-damage due to their prolonged lifespan. MSC may exert specific mechanisms of resistance to avoid loss of functional activity. We have previously shown that resistance of MSC is associated with an induction of p53 and proliferation arrest upon genotoxic damage. Hypoxia may also contribute to resistance in MSC due to the low oxygen tension in the niche. In this study we characterized the role of p53 and contribution of hypoxia in resistance of MSC to genotoxic damage. MSC exhibited increased resistance to cisplatin induced DNA-damage. This resistance was associated with a temporary G2/M cell cycle arrest, induction of p53- and p21-expression and reduced cyclin B / cdk1-levels upon subapoptotic damage. Resistance of MSC to cisplatin was increased at hypoxic conditions i. e. oxygen <0.5%. However, upon hypoxia the cisplatin-induced cell cycle arrest and expression of p53 and p21 were abrogated. MSC with shRNA-mediated p53 knock-down showed a reduced cell cycle arrest and increased cyclin B / cdk1 expression. However, this functional p53 knock down did not alter the resistance to cisplatin. In contrast to cisplatin, functional p53-knock-down increased the resistance of MSC to etoposide. We conclude that resistance of MSC to genotoxic damage is influenced by oxygen tension but is not generally dependent on p53. Thus, p53-dependent and p53-independent mechanisms of resistance are likely to contribute to the life-long functional activity of MSC in vivo. These findings indicate that hypoxia and different resistance pathways contribute to the phenotype that enables the prolonged lifespan of MSC.
Collapse
|
62
|
Santoro A, Vlachou T, Carminati M, Pelicci PG, Mapelli M. Molecular mechanisms of asymmetric divisions in mammary stem cells. EMBO Rep 2016; 17:1700-1720. [PMID: 27872203 DOI: 10.15252/embr.201643021] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/04/2016] [Accepted: 10/25/2016] [Indexed: 01/16/2023] Open
Abstract
Stem cells have the remarkable ability to undergo proliferative symmetric divisions and self-renewing asymmetric divisions. Balancing of the two modes of division sustains tissue morphogenesis and homeostasis. Asymmetric divisions of Drosophila neuroblasts (NBs) and sensory organ precursor (SOP) cells served as prototypes to learn what we consider now principles of asymmetric mitoses. They also provide initial evidence supporting the notion that aberrant symmetric divisions of stem cells could correlate with malignancy. However, transferring the molecular knowledge of circuits underlying asymmetry from flies to mammals has proven more challenging than expected. Several experimental approaches have been used to define asymmetry in mammalian systems, based on daughter cell fate, unequal partitioning of determinants and niche contacts, or proliferative potential. In this review, we aim to provide a critical evaluation of the assays used to establish the stem cell mode of division, with a particular focus on the mammary gland system. In this context, we will discuss the genetic alterations that impinge on the modality of stem cell division and their role in breast cancer development.
Collapse
Affiliation(s)
- Angela Santoro
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Thalia Vlachou
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Manuel Carminati
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | | | - Marina Mapelli
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| |
Collapse
|
63
|
Raimondi M, Marcassa E, Cataldo F, Arnandis T, Mendoza-Maldonado R, Bestagno M, Schneider C, Demarchi F. Calpain restrains the stem cells compartment in breast cancer. Cell Cycle 2016; 15:106-16. [PMID: 26771715 DOI: 10.1080/15384101.2015.1121325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
CAPNS1 is essential for the stability and function of ubiquitous CAPN1 and CAPN2. Calpain modulates by proteolytic cleavage many cellular substrates and its activity is often deregulated in cancer cells, therefore calpain inhibition has been proposed as a therapeutical strategy for a number of malignancies. Here we show that CAPNS1 depletion is coupled to impairment of MCF7 and MCF10AT cell lines growth on plate and defective architecture of mammary acini derived from MCF10A cells. In soft agar CAPNS1 depletion leads to cell growth increase in MCF7, and decrease in MCF10AT cells. In both MCF7 and MCF10AT, CAPNS1 depletion leads to the enlargement of the stem cell compartment, as demonstrated by mammosphere formation assays and evaluation of stem cell markers by means of FACS and western blot analysis. Accordingly, activation of calpain by thapsigargin treatment leads to a decrease in the stem cell reservoir. The expansion of the cancer stem cell population in CAPNS1 depleted cells is coupled to a defective shift from symmetric to asymmetric division during mammosphere growth coupled to a decrease in NUMB protein level.
Collapse
Affiliation(s)
- M Raimondi
- a L.N.C.I.B., Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie AREA Science Park - Padriciano 99 , Trieste , Italy
| | - E Marcassa
- a L.N.C.I.B., Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie AREA Science Park - Padriciano 99 , Trieste , Italy
| | - F Cataldo
- a L.N.C.I.B., Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie AREA Science Park - Padriciano 99 , Trieste , Italy
| | - T Arnandis
- a L.N.C.I.B., Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie AREA Science Park - Padriciano 99 , Trieste , Italy
| | - R Mendoza-Maldonado
- a L.N.C.I.B., Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie AREA Science Park - Padriciano 99 , Trieste , Italy
| | - M Bestagno
- c International Centre for Genetic Engineering and Biotechnology, AREA Science Park - Padriciano 99 , Trieste , Italy
| | - C Schneider
- a L.N.C.I.B., Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie AREA Science Park - Padriciano 99 , Trieste , Italy.,b Dipartimento di Scienze e Tecnologie Biomediche, Universita' degli Studi di Udine , Udine , Italy
| | - F Demarchi
- a L.N.C.I.B., Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie AREA Science Park - Padriciano 99 , Trieste , Italy
| |
Collapse
|
64
|
Li YQ, Cheng ZC, Liu SW, Aubert I, Wong CS. P53 regulates disruption of neuronal development in the adult hippocampus after irradiation. Cell Death Discov 2016; 2:16072. [PMID: 27752364 PMCID: PMC5045962 DOI: 10.1038/cddiscovery.2016.72] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/19/2016] [Indexed: 01/01/2023] Open
Abstract
Inhibition of hippocampal neurogenesis is implicated in neurocognitive dysfunction after cranial irradiation for brain tumors. How irradiation results in impaired neuronal development remains poorly understood. The Trp53 (p53) gene is known to regulate cellular DNA damage response after irradiation. Whether it has a role in disruption of late neuronal development remains unknown. Here we characterized the effects of p53 on neuronal development in adult mouse hippocampus after irradiation. Different bromodeoxyuridine incorporation paradigms and a transplantation study were used for cell fate mapping. Compared with wild-type mice, we observed profound inhibition of hippocampal neurogenesis after irradiation in mice deficient in p53 despite the absence of acute apoptosis of neuroblasts. The putative neural stem cells were apoptosis resistant after irradiation regardless of p53 genotype. Cell fate mapping using different bromodeoxyuridine incorporation paradigms revealed enhanced activation of neural stem cells and their consequential exhaustion in the absence of p53 after irradiation. Both p53-knockout and wild-type mice demonstrated similar extent of microglial activation in the hippocampus after irradiation. Impairment of neuronal differentiation of neural progenitors transplanted in irradiated hippocampus was not altered by p53 genotype of the recipient mice. We conclude that by inhibiting neural progenitor activation, p53 serves to mitigate disruption of neuronal development after irradiation independent of apoptosis and perturbation of the neural stem cell niche. These findings suggest for the first time that p53 may have a key role in late effects in brain after irradiation.
Collapse
Affiliation(s)
- Y-Q Li
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto , Toronto, ON, Canada
| | - Zw-C Cheng
- Institute of Medical Science, University of Toronto , Toronto, ON, Canada
| | - Sk-W Liu
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto , Toronto, ON, Canada
| | - I Aubert
- Department of Laboratory Medicine and Pathobiology, Sunnybrook Health Sciences Centre, University of Toronto , Toronto, ON, Canada
| | - C S Wong
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto , Toronto, ON, Canada
| |
Collapse
|
65
|
Marjault HB, Allemand I. Consequences of irradiation on adult spermatogenesis: Between infertility and hereditary risk. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 770:340-348. [DOI: 10.1016/j.mrrev.2016.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022]
|
66
|
Meng Z, Chen G, Chen J, Yang B, Yu M, Feng L, Jiang Z, Guo W, Tian W. Tumorigenicity analysis of heterogeneous dental stem cells and its self-modification for chromosome instability. Cell Cycle 2016; 14:3396-407. [PMID: 26322910 DOI: 10.1080/15384101.2015.1036204] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Heterogeneity demonstrates that stem cells are constituted by several sub-clones in various differentiation states. The heterogeneous state is maintained by cross-talk among sub-clones, thereby ensuring stem cell adaption. In this study, we investigated the roles of heterogeneity on genetic stability. Three sub-clones (DF2, DF8 and DF18) were isolated from heterogeneous dental stem cells (DSCs), and were proved to be chromosome instability (CIN) after long term expansion. Cell apoptosis were not detected in sub-clones, which exhibited strong tumorigenesis tendency, coupled with weak expression of p53 and aberrant ultra-structure. However, 3 sub-clones did not overexpress tumor related markers or induce tumorigenesis in vivo. The mixed-culture study suggested that 3-clone-mixed culturing cells (DF1) presented apparent decrease in the ratio of aneuploidy. The screening experiment further proved that 3 sub-clones functioned separately in this modification procedure but only mixed culturing all 3 sub-clones, simulated heterogeneous microenvironment, could achieve complete modification. Additionally, osteogenesis capability of 3 sub-clones was partially influenced by CIN while DSCs still kept stronger osteogenesis than sub-clones. These results suggested aberrant sub-clones isolated from heterogeneous DSCs were not tumorigenesis and could modify CIN by cross-talk among themselves, indicating that the heterogeneity played a key role in maintaining genetic stability and differentiation capability in dental stem cells.
Collapse
Affiliation(s)
- Zhaosong Meng
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,c Department of Oral and Maxillofacial Surgery ; West China School of Stomatology; Sichuan University ; Chengdu , China
| | - Guoqing Chen
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China
| | - Jinlong Chen
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,c Department of Oral and Maxillofacial Surgery ; West China School of Stomatology; Sichuan University ; Chengdu , China
| | - Bo Yang
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,c Department of Oral and Maxillofacial Surgery ; West China School of Stomatology; Sichuan University ; Chengdu , China
| | - Mei Yu
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China
| | - Lian Feng
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China
| | - Zongting Jiang
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China
| | - Weihua Guo
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,d Department of Pedodontics ; West China School of Stomatology; Sichuan University ; Chengdu , China
| | - Weidong Tian
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,c Department of Oral and Maxillofacial Surgery ; West China School of Stomatology; Sichuan University ; Chengdu , China
| |
Collapse
|
67
|
Barazzuol L, Jeggo PA. In vivo sensitivity of the embryonic and adult neural stem cell compartments to low-dose radiation. JOURNAL OF RADIATION RESEARCH 2016; 57 Suppl 1:i2-i10. [PMID: 27125639 PMCID: PMC4990107 DOI: 10.1093/jrr/rrw013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/12/2016] [Accepted: 01/17/2016] [Indexed: 05/26/2023]
Abstract
The embryonic brain is radiation-sensitive, with cognitive deficits being observed after exposure to low radiation doses. Exposure of neonates to radiation can cause intracranial carcinogenesis. To gain insight into the basis underlying these outcomes, we examined the response of the embryonic, neonatal and adult brain to low-dose radiation, focusing on the neural stem cell compartments. This review summarizes our recent findings. At E13.5-14.5 the embryonic neocortex encompasses rapidly proliferating stem and progenitor cells. Exploiting mice with a hypomorphic mutation in DNA ligase IV (Lig4(Y288C) ), we found a high level of DNA double-strand breaks (DSBs) at E14.5, which we attribute to the rapid proliferation. We observed endogenous apoptosis in Lig4(Y288C) embryos and in WT embryos following exposure to low radiation doses. An examination of DSB levels and apoptosis in adult neural stem cell compartments, the subventricular zone (SVZ) and the subgranular zone (SGZ) revealed low DSB levels in Lig4(Y288C) mice, comparable with the levels in differentiated neuronal tissues. We conclude that the adult SVZ does not incur high levels of DNA breakage, but sensitively activates apoptosis; apoptosis was less sensitively activated in the SGZ, and differentiated neuronal tissues did not activate apoptosis. P5/P15 mice showed intermediate DSB levels, suggesting that DSBs generated in the embryo can be transmitted to neonates and undergo slow repair. Interestingly, this analysis revealed a stage of high endogenous apoptosis in the neonatal SVZ. Collectively, these studies reveal that the adult neural stem cell compartment, like the embryonic counterpart, can sensitively activate apoptosis.
Collapse
Affiliation(s)
- Lara Barazzuol
- Genome Damage and Stability Centre, Life Sciences, University of Sussex, Brighton, East Sussex BN19RQ, UK
| | - Penny A Jeggo
- Genome Damage and Stability Centre, Life Sciences, University of Sussex, Brighton, East Sussex BN19RQ, UK
| |
Collapse
|
68
|
Moehrle BM, Geiger H. Aging of hematopoietic stem cells: DNA damage and mutations? Exp Hematol 2016; 44:895-901. [PMID: 27402537 DOI: 10.1016/j.exphem.2016.06.253] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 06/27/2016] [Indexed: 12/15/2022]
Abstract
Aging in the hematopoietic system and the stem cell niche contributes to aging-associated phenotypes of hematopoietic stem cells (HSCs), including leukemia and aging-associated immune remodeling. Among others, the DNA damage theory of aging of HSCs is well established, based on the detection of a significantly larger amount of γH2AX foci and a higher tail moment in the comet assay, both initially thought to be associated with DNA damage in aged HSCs compared with young cells, and bone marrow failure in animals devoid of DNA repair factors. Novel data on the increase in and nature of DNA mutations in the hematopoietic system with age, the quality of the DNA damage response in aged HSCs, and the nature of γH2AX foci question a direct link between DNA damage and the DNA damage response and aging of HSCs, and rather favor changes in epigenetics, splicing-factors or three-dimensional architecture of the cell as major cell intrinsic factors of HSCs aging. Aging of HSCs is also driven by a strong contribution of aging of the niche. This review discusses the DNA damage theory of HSC aging in the light of these novel mechanisms of aging of HSCs.
Collapse
Affiliation(s)
| | - Hartmut Geiger
- Institute for Molecular Medicine, Ulm University, Ulm, Germany; Aging Research Center, Ulm University, Ulm, Germany; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| |
Collapse
|
69
|
Li YQ, Cheng Z, Wong S. Differential Apoptosis Radiosensitivity of Neural Progenitors in Adult Mouse Hippocampus. Int J Mol Sci 2016; 17:ijms17060970. [PMID: 27331809 PMCID: PMC4926502 DOI: 10.3390/ijms17060970] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 06/01/2016] [Accepted: 06/13/2016] [Indexed: 12/22/2022] Open
Abstract
Mammalian tissue-specific stem cells and progenitors demonstrate differential DNA damage response. Neural progenitors in dentate gyrus of the hippocampus are known to undergo apoptosis after irradiation. Using a mouse model of hippocampal neuronal development, we characterized the apoptosis sensitivity of the different neural progenitor subpopulations in adult mouse dentate gyrus after irradiation. Two different bromodeoxyuridine incorporation paradigms were used for cell fate mapping. We identified two apoptosis sensitive neural progenitor subpopulations after irradiation. The first represented non-proliferative and non-newborn neuroblasts and immature neurons that expressed doublecortin, calretinin or both. The second consisted of proliferative intermediate neural progenitors. The putative radial glia-like neural stem cells or type-1 cells, regardless of proliferation status, were apoptosis resistant after irradiation. There was no evidence of radiation-induced apoptosis in the absence of the Trp53 (p53) gene but absence of Cdkn1a (p21) did not alter the apoptotic response. Upregulation of nuclear p53 was observed in neuroblasts after irradiation. We conclude that adult hippocampal neural progenitors may demonstrate differential p53-dependent apoptosis sensitivity after irradiation.
Collapse
Affiliation(s)
- Yu-Qing Li
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada.
| | - Zoey Cheng
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Shun Wong
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Departments of Radiation Oncology and Medical Biophysics, University of Toronto, Toronto, ON M4N 3M5, Canada.
| |
Collapse
|
70
|
|
71
|
DNA Damage Response in Hematopoietic Stem Cell Ageing. GENOMICS PROTEOMICS & BIOINFORMATICS 2016; 14:147-154. [PMID: 27221660 PMCID: PMC4936660 DOI: 10.1016/j.gpb.2016.04.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/20/2016] [Accepted: 04/24/2016] [Indexed: 12/30/2022]
Abstract
Maintenance of tissue-specific stem cells is vital for organ homeostasis and organismal longevity. Hematopoietic stem cells (HSCs) are the most primitive cell type in the hematopoietic system. They divide asymmetrically and give rise to daughter cells with HSC identity (self-renewal) and progenitor progenies (differentiation), which further proliferate and differentiate into full hematopoietic lineages. Mammalian ageing process is accompanied with abnormalities in the HSC self-renewal and differentiation. Transcriptional changes and epigenetic modulations have been implicated as the key regulators in HSC ageing process. The DNA damage response (DDR) in the cells involves an orchestrated signaling pathway, consisting of cell cycle regulation, cell death and senescence, transcriptional regulation, as well as chromatin remodeling. Recent studies employing DNA repair-deficient mouse models indicate that DDR could intrinsically and extrinsically regulate HSC maintenance and play important roles in tissue homeostasis of the hematopoietic system. In this review, we summarize the current understanding of how the DDR determines the HSC fates and finally contributes to organismal ageing.
Collapse
|
72
|
Tosoni D, Zecchini S, Coazzoli M, Colaluca I, Mazzarol G, Rubio A, Caccia M, Villa E, Zilian O, Di Fiore PP, Pece S. The Numb/p53 circuitry couples replicative self-renewal and tumor suppression in mammary epithelial cells. J Cell Biol 2016; 211:845-62. [PMID: 26598619 PMCID: PMC4657167 DOI: 10.1083/jcb.201505037] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The cell fate determinant Numb orchestrates tissue morphogenesis and patterning in developmental systems. In the human mammary gland, Numb is a tumor suppressor and regulates p53 levels. However, whether this function is linked to its role in fate determination remains unclear. Here, by exploiting an ex vivo system, we show that at mitosis of purified mammary stem cells (SCs), Numb ensures the asymmetric outcome of self-renewing divisions by partitioning into the progeny that retains the SC identity, where it sustains high p53 activity. Numb also controls progenitor maturation. At this level, Numb loss associates with the epithelial-to-mesenchymal transition and results in differentiation defects and reacquisition of stemness features. The mammary gland of Numb-knockout mice displays an expansion of the SC compartment, associated with morphological alterations and tumorigenicity in orthotopic transplants. This is because of low p53 levels and can be inhibited by restoration of Numb levels or p53 activity, which results in successful SC-targeted treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Alicia Rubio
- Istituto Europeo di Oncologia, 20141 Milan, Italy
| | | | | | | | - Pier Paolo Di Fiore
- Istituto Europeo di Oncologia, 20141 Milan, Italy Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy Fondazione Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy
| | - Salvatore Pece
- Istituto Europeo di Oncologia, 20141 Milan, Italy Fondazione Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy
| |
Collapse
|
73
|
Pérez LM, Suárez J, Bernal A, de Lucas B, San Martin N, Gálvez BG. Obesity-driven alterations in adipose-derived stem cells are partially restored by weight loss. Obesity (Silver Spring) 2016; 24:661-9. [PMID: 26833860 DOI: 10.1002/oby.21405] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 10/13/2015] [Accepted: 10/19/2015] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The therapeutic potential of adipose-derived stem cells (ASCs) is reduced by various stress-inducing conditions that affect tissue homeostasis such as diabetes, aging, and obesity. Previous works have provided evidence of negative effects of obesity on ASC populations, but it is unclear whether this persists after a weight loss. This study evaluated whether weight loss can restore the attenuated properties found in ASCs derived from populations with obesity (oASCs). METHODS In vitro functional analyses were performed to investigate the possible recovery properties in mouse oASCs. Using ASCs isolated from subcutaneous tissue from formerly obese mice (dASCs) and control mice (cASCs), cell proliferation, viability, and some regenerative properties in these cells were analyzed compared with oASCs to evaluate the functional cell state. RESULTS Cell proliferation, viability, and some regenerative properties are strengthened in dASCs and cASCs compared with oASCs. Nevertheless, metabolic analysis reveals a mitochondrial load misbalance and function leading to impaired respiration in dASCs. CONCLUSIONS This study demonstrates that an initial obese environment triggers a detrimental state in ASCs that is not completely recovered after weight loss.
Collapse
Affiliation(s)
- Laura M Pérez
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Javier Suárez
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Aurora Bernal
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz de Lucas
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Nuria San Martin
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz G Gálvez
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- School of Doctorate and Research, European University, Madrid, Spain
| |
Collapse
|
74
|
Cheng NC, Hsieh TY, Lai HS, Young TH. High glucose-induced reactive oxygen species generation promotes stemness in human adipose-derived stem cells. Cytotherapy 2016; 18:371-83. [PMID: 26780864 DOI: 10.1016/j.jcyt.2015.11.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Adipose-derived stem cells (ASCs) represent an important source of cell therapy to treat diabetic complications. However, hyperglycemia may alter several cellular functions, so the present study aimed to investigate the influence of a diabetic environment on the stemness and differentiation capabilities of ASCs. METHODS Human ASCs were obtained from subcutaneous adipose tissues of diabetic (dASCs) and nondiabetic donors (nASCs) and characterized. To reproduce an in vitro hyperglycemia environment, the nASCs were also cultured under prolonged high-glucose (HG; 4.5 g/L) or low-glucose (LG; 1.0 g/L) conditions. RESULTS The expression of cell surface markers in dASCs and nASC was similar and characteristic of mesenchymal stem cells. Although dASCs or HG-treated nASCs exhibited decreased proliferation, enhanced expression of the pluripotent markers Sox-2, Oct-4, and Nanog was observed. Moreover, HG-treated nASCs exhibited decreased cell migration, enhanced senescence, and significantly higher intracellular reactive oxygen species (ROS), whereas their adipogenic and osteogenic differentiation capacities remained comparable to LG-treated cells. With antioxidant treatment, HG-treated nASCs showed improved cell proliferative activity without stemness enhancement. This HG-induced biological response was associated with ROS-mediated AKT attenuation. When cultured in an appropriate induction medium, the HG-treated nASCs and dASCs exhibited enhanced potential of transdifferentiation into neuron-like cells. DISCUSSION Despite lower proliferative activity and higher senescence in a diabetic environment, ASCs also exhibit enhanced stemness and neurogenic transdifferentiation potential via a ROS-mediated mechanism. The information is important for future application of autologous ASCs in diabetic patients.
Collapse
Affiliation(s)
- Nai-Chen Cheng
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsung-Yu Hsieh
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Hong-Shiee Lai
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Tai-Horng Young
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
75
|
Abstract
A major barrier to achieving durable remission and a definitive cure in oncology patients is the emergence of tumor resistance, a common outcome of different disease types, and independent from the therapeutic approach undertaken. In recent years, subpopulations of slow-cycling cells endowed with enhanced tumorigenic potential and multidrug resistance have been isolated in different tumors, and mounting experimental evidence suggests these resistant cells are responsible for tumor relapse. An in-depth metabolic characterization of resistant tumor stem cells revealed that they rely more on mitochondrial respiration and less on glycolysis than other tumor cells, a finding that challenges the assumption that tumors have a primarily glycolytic metabolism and defective mitochondria. The demonstration of a metabolic program in resistant tumorigenic cells that may be present in the majority of tumors has important therapeutic implications and is a critical consideration as we address the challenge of identifying new vulnerabilities that might be exploited therapeutically.
Collapse
Affiliation(s)
- Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
76
|
Moehrle BM, Nattamai K, Brown A, Florian MC, Ryan M, Vogel M, Bliederhaeuser C, Soller K, Prows DR, Abdollahi A, Schleimer D, Walter D, Milsom MD, Stambrook P, Porteus M, Geiger H. Stem Cell-Specific Mechanisms Ensure Genomic Fidelity within HSCs and upon Aging of HSCs. Cell Rep 2015; 13:2412-2424. [PMID: 26686632 DOI: 10.1016/j.celrep.2015.11.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/13/2015] [Accepted: 11/08/2015] [Indexed: 01/22/2023] Open
Abstract
Whether aged hematopoietic stem and progenitor cells (HSPCs) have impaired DNA damage repair is controversial. Using a combination of DNA mutation indicator assays, we observe a 2- to 3-fold increase in the number of DNA mutations in the hematopoietic system upon aging. Young and aged hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) do not show an increase in mutation upon irradiation-induced DNA damage repair, and young and aged HSPCs respond very similarly to DNA damage with respect to cell-cycle checkpoint activation and apoptosis. Both young and aged HSPCs show impaired activation of the DNA-damage-induced G1-S checkpoint. Induction of chronic DNA double-strand breaks by zinc-finger nucleases suggests that HSPCs undergo apoptosis rather than faulty repair. These data reveal a protective mechanism in both the young and aged hematopoietic system against accumulation of mutations in response to DNA damage.
Collapse
Affiliation(s)
- Bettina M Moehrle
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Kalpana Nattamai
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Andreas Brown
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Maria C Florian
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Marnie Ryan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Mona Vogel
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | | | - Karin Soller
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Daniel R Prows
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Amir Abdollahi
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Molecular and Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), 69120 Heidelberg, Germany
| | - David Schleimer
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Dagmar Walter
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine gGmbH (HI-STEM), 69120 Heidelberg, Germany
| | - Michael D Milsom
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine gGmbH (HI-STEM), 69120 Heidelberg, Germany; Deutsches Krebsforschungszentrum (DKFZ), Division of Stem Cells and Cancer, Experimental Hematology Group, 69120 Heidelberg, Germany
| | - Peter Stambrook
- Department of Molecular Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Matthew Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Hartmut Geiger
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
77
|
Mieloch AA, Suchorska WM. The concept of radiation-enhanced stem cell differentiation. Radiol Oncol 2015; 49:209-16. [PMID: 26401125 PMCID: PMC4577216 DOI: 10.1515/raon-2015-0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/05/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Efficient stem cell differentiation is considered to be the holy grail of regenerative medicine. Pursuing the most productive method of directed differentiation has been the subject of numerous studies, resulting in the development of many effective protocols. However, the necessity for further improvement in differentiation efficiency remains. This review contains a description of molecular processes underlying the response of stem cells to ionizing radiation, indicating its potential application in differentiation procedures. In the first part, the radiation-induced damage response in various types of stem cells is described. Second, the role of the p53 protein in embryonic and adult stem cells is highlighted. Last, the hypothesis on the mitochondrial involvement in stem cell development including its response to ionizing radiation is presented. CONCLUSIONS In summary, despite the many threats of ionizing radiation concerning genomic instability, subjecting cells to the appropriate dosage of ionizing radiation may become a useful method for enhancing directed differentiation in certain stem cell types.
Collapse
Affiliation(s)
- Adam A. Mieloch
- Radiobiology Laboratory, Department of Medical Physics, The Greater Poland Cancer Centre
| | - Wiktoria M. Suchorska
- Radiobiology Laboratory, Department of Medical Physics, The Greater Poland Cancer Centre
| |
Collapse
|
78
|
Chang CH, Zhang M, Rajapakshe K, Coarfa C, Edwards D, Huang S, Rosen JM. Mammary Stem Cells and Tumor-Initiating Cells Are More Resistant to Apoptosis and Exhibit Increased DNA Repair Activity in Response to DNA Damage. Stem Cell Reports 2015; 5:378-91. [PMID: 26300228 PMCID: PMC4618454 DOI: 10.1016/j.stemcr.2015.07.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 01/05/2023] Open
Abstract
Adult stem cells and tumor-initiating cells (TICs) often employ different mechanisms of DNA damage response (DDR) as compared to other tissue cell types. However, little is known about how mammary stem cells (MaSCs) and mammary TICs respond to DNA damage. Using the mouse mammary gland and syngeneic p53-null tumors as models, we investigated the molecular and physiological consequences of DNA damage in wild-type MaSCs, p53-null MaSCs, and p53-null TICs. We showed that wild-type MaSCs and basal cells are more resistant to apoptosis and exhibit increased non-homologous end joining (NHEJ) activity. Loss of p53 in mammary epithelium affected both cell-cycle regulation and DNA repair efficiency. In p53-null tumors, we showed that TICs are more resistant to ionizing radiation (IR) due to decreased apoptosis, elevated NHEJ activity, and more-rapid DNA repair. These results have important implications for understanding DDR mechanisms involved in both tumorigenesis and therapy resistance. MaSCs are more resistant to apoptosis and exhibit increased NHEJ activity Loss of p53 in MECs impairs cell-cycle regulation and DNA repair efficiency TICs exhibit decreased apoptosis, increased DNA repair efficiency, and NHEJ activity TICs are highly proliferative and exhibit improper cell-cycle regulation after IR
Collapse
Affiliation(s)
- Chi-Hsuan Chang
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mei Zhang
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dean Edwards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeffrey M Rosen
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
79
|
Song J, Kang SM, Kim E, Kim CH, Song HT, Lee JE. Adiponectin receptor-mediated signaling ameliorates cerebral cell damage and regulates the neurogenesis of neural stem cells at high glucose concentrations: an in vivo and in vitro study. Cell Death Dis 2015; 6:e1844. [PMID: 26247729 PMCID: PMC4558511 DOI: 10.1038/cddis.2015.220] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/03/2015] [Accepted: 07/06/2015] [Indexed: 01/06/2023]
Abstract
In the central nervous system (CNS), hyperglycemia leads to neuronal damage and cognitive decline. Recent research has focused on revealing alterations in the brain in hyperglycemia and finding therapeutic solutions for alleviating the hyperglycemia-induced cognitive dysfunction. Adiponectin is a protein hormone with a major regulatory role in diabetes and obesity; however, its role in the CNS has not been studied yet. Although the presence of adiponectin receptors has been reported in the CNS, adiponectin receptor-mediated signaling in the CNS has not been investigated. In the present study, we investigated adiponectin receptor (AdipoR)-mediated signaling in vivo using a high-fat diet and in vitro using neural stem cells (NSCs). We showed that AdipoR1 protects cell damage and synaptic dysfunction in the mouse brain in hyperglycemia. At high glucose concentrations in vitro, AdipoR1 regulated the survival of NSCs through the p53/p21 pathway and the proliferation- and differentiation-related factors of NSCs via tailless (TLX). Hence, we suggest that further investigations are necessary to understand the cerebral AdipoR1-mediated signaling in hyperglycemic conditions, because the modulation of AdipoR1 might alleviate hyperglycemia-induced neuropathogenesis.
Collapse
Affiliation(s)
- J Song
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - S M Kang
- 1] Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, South Korea [2] BK21 Plus Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - E Kim
- Department of Psychiatry, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - C-H Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - H-T Song
- Department of Diagnostic Radiology, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - J E Lee
- 1] Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, South Korea [2] BK21 Plus Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul 120-752, South Korea
| |
Collapse
|
80
|
Abstract
SIGNIFICANCE Epigenetic inactivation of pivotal genes involved in cell growth is a hallmark of human pathologies, in particular cancer. Histone acetylation balance obtained through opposing actions of histone deacetylases (HDACs) and histone acetyltransferases is one epigenetic mechanism controlling gene expression and is, thus, associated with disease etiology and progression. Interfering pharmacologically with HDAC activity can correct abnormalities in cell proliferation, migration, vascularization, and death. RECENT ADVANCES Histone deacetylase inhibitors (HDACi) represent a new class of cytostatic agents that interfere with the function of HDACs and are able to increase gene expression by indirectly inducing histone acetylation. Several HDACi, alone or in combination with DNA-demethylating agents, chemopreventive, or classical chemotherapeutic drugs, are currently being used in clinical trials for solid and hematological malignancies, and are, thus, promising candidates for cancer therapy. CRITICAL ISSUES (i) Non-specific (off-target) HDACi effects due to activities unassociated with HDAC inhibition. (ii) Advantages/disadvantages of non-selective or isoform-directed HDACi. (iii) Limited number of response-predictive biomarkers. (iv) Toxicity leading to dysfunction of critical biological processes. FUTURE DIRECTIONS Selective HDACi could achieve enhanced clinical utility by reducing or eliminating the serious side effects associated with current first-generation non-selective HDACi. Isoform-selective and pan-HDACi candidates might benefit from the identification of biomarkers, enabling better patient stratification and prediction of response to treatment.
Collapse
Affiliation(s)
- Rosaria Benedetti
- 1 Department of Biochemistry, Biophysics, and General Pathology, Seconda Università degli Studi di Napoli , Napoli, Italy
| | - Mariarosaria Conte
- 1 Department of Biochemistry, Biophysics, and General Pathology, Seconda Università degli Studi di Napoli , Napoli, Italy
| | - Lucia Altucci
- 1 Department of Biochemistry, Biophysics, and General Pathology, Seconda Università degli Studi di Napoli , Napoli, Italy .,2 Istituto di Genetica e Biofisica "Adriano Buzzati-Traverso," Napoli, Italy
| |
Collapse
|
81
|
Colla S, Ong DST, Ogoti Y, Marchesini M, Mistry NA, Clise-Dwyer K, Ang SA, Storti P, Viale A, Giuliani N, Ruisaard K, Ganan Gomez I, Bristow CA, Estecio M, Weksberg DC, Ho YW, Hu B, Genovese G, Pettazzoni P, Multani AS, Jiang S, Hua S, Ryan MC, Carugo A, Nezi L, Wei Y, Yang H, D'Anca M, Zhang L, Gaddis S, Gong T, Horner JW, Heffernan TP, Jones P, Cooper LJN, Liang H, Kantarjian H, Wang YA, Chin L, Bueso-Ramos C, Garcia-Manero G, DePinho RA. Telomere dysfunction drives aberrant hematopoietic differentiation and myelodysplastic syndrome. Cancer Cell 2015; 27:644-57. [PMID: 25965571 PMCID: PMC4596059 DOI: 10.1016/j.ccell.2015.04.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 01/31/2015] [Accepted: 04/13/2015] [Indexed: 12/14/2022]
Abstract
Myelodysplastic syndrome (MDS) risk correlates with advancing age, therapy-induced DNA damage, and/or shorter telomeres, but whether telomere erosion directly induces MDS is unknown. Here, we provide the genetic evidence that telomere dysfunction-induced DNA damage drives classical MDS phenotypes and alters common myeloid progenitor (CMP) differentiation by repressing the expression of mRNA splicing/processing genes, including SRSF2. RNA-seq analyses of telomere dysfunctional CMP identified aberrantly spliced transcripts linked to pathways relevant to MDS pathogenesis such as genome stability, DNA repair, chromatin remodeling, and histone modification, which are also enriched in mouse CMP haploinsufficient for SRSF2 and in CD34(+) CMML patient cells harboring SRSF2 mutation. Together, our studies establish an intimate link across telomere biology, aberrant RNA splicing, and myeloid progenitor differentiation.
Collapse
Affiliation(s)
- Simona Colla
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Derrick Sek Tong Ong
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yamini Ogoti
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matteo Marchesini
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nipun A Mistry
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sonny A Ang
- Department of Pediatric Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paola Storti
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Hematology, Department of Clinical and Experimental Medicine, University of Parma, 43126 Parma, Italy
| | - Andrea Viale
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicola Giuliani
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, 43126 Parma, Italy
| | - Kathryn Ruisaard
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Irene Ganan Gomez
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher A Bristow
- Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marcos Estecio
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | - David C Weksberg
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Wing Ho
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Baoli Hu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Giannicola Genovese
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Piergiorgio Pettazzoni
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Asha S Multani
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shan Jiang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sujun Hua
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Alessandro Carugo
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Luigi Nezi
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yue Wei
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui Yang
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marianna D'Anca
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Li Zhang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sarah Gaddis
- Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | - Ting Gong
- Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | - James W Horner
- Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Timothy P Heffernan
- Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Philip Jones
- Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Laurence J N Cooper
- Department of Pediatric Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Y Alan Wang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lynda Chin
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Carlos Bueso-Ramos
- Department of Hematopathology, University of Texas MD Cancer Center, Houston, TX 77030, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
82
|
The functional interplay between the t(9;22)-associated fusion proteins BCR/ABL and ABL/BCR in Philadelphia chromosome-positive acute lymphatic leukemia. PLoS Genet 2015; 11:e1005144. [PMID: 25919613 PMCID: PMC4412790 DOI: 10.1371/journal.pgen.1005144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 03/15/2015] [Indexed: 12/20/2022] Open
Abstract
The hallmark of Philadelphia chromosome positive (Ph+) leukemia is the BCR/ABL kinase, which is successfully targeted by selective ATP competitors. However, inhibition of BCR/ABL alone is unable to eradicate Ph+ leukemia. The t(9;22) is a reciprocal translocation which encodes not only for the der22 (Philadelphia chromosome) related BCR/ABL, but also for der9 related ABL/BCR fusion proteins, which can be detected in 65% of patients with chronic myeloid leukemia (CML) and 100% of patients with Ph+ acute lymphatic leukemia (ALL). ABL/BCRs are oncogenes able to influence the lineage commitment of hematopoietic progenitors. Aim of this study was to further disclose the role of p96ABL/BCR for the pathogenesis of Ph+ ALL. The co-expression of p96ABL/BCR enhanced the kinase activity and as a consequence, the transformation potential of p185BCR/ABL. Targeting p96ABL/BCR by RNAi inhibited growth of Ph+ ALL cell lines and Ph+ ALL patient-derived long-term cultures (PD-LTCs). Our in vitro and in vivo stem cell studies further revealed a functional hierarchy of p96ABL/BCR and p185BCR/ABL in hematopoietic stem cells. Co-expression of p96ABL/BCR abolished the capacity of p185BCR/ABL to induce a CML-like disease and led to the induction of ALL. Taken together our here presented data reveal an important role of p96ABL/BCR for the pathogenesis of Ph+ ALL. The t(9;22) is a reciprocal translocation, which causes chronic myeloid leukemia (CML) and a subset of high risk acute lymphatic leukemia (ALL). The derivative chromosome 22 is the so called Philadelphia chromosome (Ph) which encodes the BCR/ABL kinase. Targeting BCR/ABL by selective ATP competitors, such as imatinib or nilotinib, is a well validated therapeutic concept, but unable to definitively eradicate the disease. Little is known about the role of the fusion protein encoded by the reciprocal derivative chromosome 9, the ABL/BCR. In models of Ph+ ALL we show that the functional interplay between ABL/BCR and BCR/ABL not only increases the transformation potential of BCR/ABL but is also indispensable for the growth and survival of Ph+ ALL leukemic cells. The presence of ABL/BCR changed the phenotype of the leukemia most likely due to its capacity to influence the stem cell population as shown by our in vivo data. Taken together our here presented data reveal an important role of p96ABL/BCR for the pathogenesis of Ph+ ALL.
Collapse
|
83
|
Falzacappa MVV, Ronchini C, Faretta M, Iacobucci I, Di Rorà AGL, Martinelli G, Meyer LH, Debatin KM, Orecchioni S, Bertolini F, Pelicci PG. The Combination of the PARP Inhibitor Rucaparib and 5FU Is an Effective Strategy for Treating Acute Leukemias. Mol Cancer Ther 2015; 14:889-98. [PMID: 25667168 DOI: 10.1158/1535-7163.mct-14-0276] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 02/02/2015] [Indexed: 11/16/2022]
Abstract
The existing treatments to cure acute leukemias seem to be nonspecific and suboptimal for most patients, drawing attention to the need of new therapeutic strategies. In the last decade the anticancer potential of poly ADP-ribose polymerase (PARP) inhibitors became apparent and now several PARP inhibitors are being developed to treat various malignancies. So far, the usage of PARP inhibitors has been mainly focused on the treatment of solid tumors and not too much about their efficacy on leukemias is known. In this study we test, for the first time on leukemic cells, a combined therapy that associates the conventional chemotherapeutic agent fluorouracil (5FU), used as a source of DNA damage, and a PARP inhibitor, rucaparib. We demonstrate the efficacy and the specificity of this combined therapy in killing both acute myeloid leukemia and acute lymphoid leukemia cells in vitro and in vivo. We clearly show that the inhibition of DNA repair induced by rucaparib is synthetic lethal with the DNA damage caused by 5FU in leukemic cells. Therefore, we propose a new therapeutic strategy able to enhance the cytotoxic effect of DNA-damaging agents in leukemia cells via inhibiting the repair of damaged DNA.
Collapse
Affiliation(s)
| | - Chiara Ronchini
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia, Milan, Italy
| | - Mario Faretta
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Ilaria Iacobucci
- Institute of Hematology "L. e A. Seràgnoli," Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Andrea Ghelli Luserna Di Rorà
- Institute of Hematology "L. e A. Seràgnoli," Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Giovanni Martinelli
- Institute of Hematology "L. e A. Seràgnoli," Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Lüder Hinrich Meyer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, European Institute of Oncology, Milan, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology, Milan, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy. Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
84
|
Li RF, Feng YQ, Chen JH, Ge LT, Xiao SY, Zuo XL. Naringenin suppresses K562 human leukemia cell proliferation and ameliorates Adriamycin-induced oxidative damage in polymorphonuclear leukocytes. Exp Ther Med 2015; 9:697-706. [PMID: 25667616 PMCID: PMC4316947 DOI: 10.3892/etm.2015.2185] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 12/04/2014] [Indexed: 12/13/2022] Open
Abstract
Treatments for leukemia remain unsatisfactory. Conventional chemotherapy agents that aim to kill tumor cells may also damage normal cells and thus result in severe side-effects. Naringenin, a natural polyphenolic compound with antioxidant effects, has been revealed to have significant antitumor effects with low toxicity in preliminary studies. Thus, it is considered as one of the most promising flavonoids in the treatment of leukemia. In the present study, the effects of naringenin on the K562 human leukemia cell line and the underlying mechanisms were explored in vitro. In addition, human peripheral blood polymorphonuclear leukocytes (PMNs) were used as a normal control in order to evaluate the effects of naringenin on normal granulocytes and in the mediation of Adriamycin (ADM)-induced oxidative damage. The results revealed that K562 proliferation was significantly inhibited by naringenin in a time- and concentration-dependent manner; however, minimal cytotoxic effects were observed in PMNs when naringenin was used at concentrations <400 μmol/l. Morphological changes indicative of apoptosis were observed in naringenin-treated K562 cells. Flow cytometric analysis indicated that the K562 cells were arrested in the G0/G1 phase of the cell cycle with a significantly upregulated rate of apoptosis. Furthermore, in the naringenin-treated K562 cells, the labeling index of proliferating cell nuclear antigen was observed to be increased by immunochemical staining, the mRNA and protein expression levels of p21/WAF1 were strongly upregulated in reverse transcription-polymerase chain reaction and western blot analyses, whereas p53 gene expression was not significantly changed. In PMNs to which naringenin (50~80 μmol/l) was added 1 h subsequent to ADM, the cell damage induced by ADM was significantly reduced, coincident with reductions in the levels of reactive oxygen species (ROS) and malondialdehyde (MDA) and increases in the activity of superoxide dismutase and glutathione peroxidase. However, the cytotoxic effect of ADM in K562 cells was not significantly altered by naringenin, and the oxidative stress indices in K562 cells remained stable. In conclusion, the present study revealed the promising value of naringenin in leukemia treatment. Naringenin demonstrated a significant inhibitory effect on the growth of K562 cells but not on normal PMNs. Furthermore, naringenin protected PMNs from ADM-induced oxidative damage at low concentrations. Cell cycle arrest and apoptosis-inducing effects, achieved through p53-independent p21/WAF1 upregulation, are likely to be the mechanism of the antileukemic effects of naringenin, and the protective effect against ADM chemotherapy-induced damage in PMNs may be due to the antioxidant capability of this agent at low concentrations.
Collapse
Affiliation(s)
- Rui-Fang Li
- Department of Neurology, Hubei Zhongshan Hospital, Wuhan, Hubei 430033, P.R. China ; Department of Pathology, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Ying-Qian Feng
- Department of Endocrinology, Weapon Industry 521 Hospital, Xi'an, Shaanxi 710065, P.R. China
| | - Jun-Hui Chen
- Department of Science and Education, Hubei Zhongshan Hospital, Wuhan, Hubei 430033, P.R. China
| | - Lin-Tong Ge
- Department of Neurology, Hubei Zhongshan Hospital, Wuhan, Hubei 430033, P.R. China
| | - Shu-Yuan Xiao
- Department of Pathology, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Xue-Lan Zuo
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
85
|
JIAO YANG, LIU CHANG, CUI FENGMEI, XU JIAYING, TONG JIAN, QI XIAOFEI, WANG LILI, ZHU WEI. Long intergenic non-coding RNA induced by X-ray irradiation regulates DNA damage response signaling in the human bronchial epithelial BEAS-2B cell line. Oncol Lett 2015; 9:169-176. [PMID: 25435953 PMCID: PMC4247013 DOI: 10.3892/ol.2014.2622] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 09/26/2014] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been regarded as the primary genetic regulators of several important biological processes. However, the biological functions of lncRNAs in radiation-induced lung damage remain largely unknown. The present study aimed to investigate the potential effects of lncRNAs on radiation-induced lung injury (RILI). Female C57BL/6 mice were exposed to 12 Gy single doses of total body irradiation (TBI). LncRNA microarray screening was conducted at 24 h post-irradiation (IR) to investigate the differentially-expressed lncRNAs during RILI. Following the subsequent bioinformatics analysis and reverse transcription-polymerase chain reaction (RT-PCR) validation, one of the verified differentially-expressed long intergenic radiation-responsive ncRNAs (LIRRs), LIRR1, was selected for further functional study. The normal human bronchial epithelial BEAS-2B cell line was used as the cell model. The recombinant eukaryotic expression vector for the lncRNA was designed, constructed and transfected using lipofectamine. RT-PCR, clonogenic and flow cytometry assays, immunofluorescence detection and western blot analysis were performed to reveal the role of the lncRNA in the radiosensitivity regulation of the RILI target cells. In lung tissues 24 h after 12 Gy TBI, six of the identified differentially-expressed LIRRs near the coding genes were validated using quantitative (q)PCR. The upregulation of two LIRRs was observed and confirmed using qPCR. LIRR1 was chosen for further functional study. Following the stable transfection of LIRR1, identified through G418 screening, increased radiosensitivity, evident cell cycle G1 phase arrest and increased γ-H2AX foci formation were observed in the bronchial epithelial BEAS-2B cell line subsequent to IR. LIRR1 overexpression also led to decreased expression of the KU70, KU80 and RAD50 DNA repair proteins, marked activation of p53, decreased mouse double minute 2 homolog (MDM2) expression, and substantially induced p21 and suppressed cyclin-dependent kinase 2 in BEAS-2B following IR. Subsequent to the use of Pifithrin-α, a specific inhibitor of p53 activation, increased MDM2 expression was observed in the LIRR1-overexpressing cells, suggesting that LIRR1 could mediate the DNA damage response (DDR) signaling in a p53-dependent manner. The present study provides a novel mechanism for RILI, using the concept of lncRNAs.
Collapse
Affiliation(s)
- YANG JIAO
- Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - CHANG LIU
- Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - FENG-MEI CUI
- Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - JIA-YING XU
- Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - JIAN TONG
- School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - XIAO-FEI QI
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - LI-LI WANG
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - WEI ZHU
- Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
86
|
De Stefano I, Tanno B, Giardullo P, Leonardi S, Pasquali E, Antonelli F, Tanori M, Casciati A, Pazzaglia S, Saran A, Mancuso M. The Patched 1 tumor-suppressor gene protects the mouse lens from spontaneous and radiation-induced cataract. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:85-95. [PMID: 25452120 DOI: 10.1016/j.ajpath.2014.09.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/17/2014] [Accepted: 09/04/2014] [Indexed: 11/16/2022]
Abstract
Age-related cataract is the most common cause of visual impairment. Moreover, traumatic cataracts form after injury to the eye, including radiation damage. We report herein that sonic hedgehog (Shh) signaling plays a key role in cataract development and in normal lens response to radiation injury. Mice heterozygous for Patched 1 (Ptch1), the Shh receptor and negative regulator of the pathway, develop spontaneous cataract and are highly susceptible to cataract induction by exposure to ionizing radiation in early postnatal age, when lens epithelial cells undergo rapid expansion in the lens epithelium. Neonatally irradiated and control Ptch1(+/-) mice were compared for markers of progenitors, Shh pathway activation, and epithelial-to-mesenchymal transition (EMT). Molecular analyses showed increased expression of the EMT-related transforming growth factor β/Smad signaling pathway in the neonatally irradiated lens, and up-regulation of mesenchymal markers Zeb1 and Vim. We further show a link between proliferation and the stemness property of lens epithelial cells, controlled by Shh. Our results suggest that Shh and transforming growth factor β signaling cooperate to promote Ptch1-associated cataract development by activating EMT, and that the Nanog marker of pluripotent cells may act as the primary transcription factor on which both signaling pathways converge after damage. These findings highlight a novel function of Shh signaling unrelated to cancer and provide a new animal model to investigate the molecular pathogenesis of cataract formation.
Collapse
Affiliation(s)
- Ilaria De Stefano
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy
| | - Barbara Tanno
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Paola Giardullo
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy
| | - Simona Leonardi
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Emanuela Pasquali
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Francesca Antonelli
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Mirella Tanori
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Arianna Casciati
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Simonetta Pazzaglia
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Anna Saran
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy.
| | - Mariateresa Mancuso
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy.
| |
Collapse
|
87
|
Tang J, Fernandez-Garcia I, Vijayakumar S, Martinez-Ruis H, Illa-Bochaca I, Nguyen DH, Mao JH, Costes SV, Barcellos-Hoff MH. Irradiation of juvenile, but not adult, mammary gland increases stem cell self-renewal and estrogen receptor negative tumors. Stem Cells 2014; 32:649-61. [PMID: 24038768 DOI: 10.1002/stem.1533] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 06/30/2013] [Accepted: 07/24/2013] [Indexed: 02/06/2023]
Abstract
Children exposed to ionizing radiation have a substantially greater breast cancer risk than adults; the mechanism for this strong age dependence is not known. Here we show that pubertal murine mammary glands exposed to sparsely or densely ionizing radiation exhibit enrichment of mammary stem cell and Notch pathways, increased mammary repopulating activity indicative of more stem cells, and propensity to develop estrogen receptor (ER) negative tumors thought to arise from stem cells. We developed a mammary lineage agent-based model (ABM) to evaluate cell inactivation, self-renewal, or dedifferentiation via epithelial-mesenchymal transition (EMT) as mechanisms by which radiation could increase stem cells. ABM rejected cell inactivation and predicted increased self-renewal would only affect juveniles while dedifferentiation could act in both juveniles and adults. To further test self-renewal versus dedifferentiation, we used the MCF10A human mammary epithelial cell line, which recapitulates ductal morphogenesis in humanized fat pads, undergoes EMT in response to radiation and transforming growth factor β (TGFβ) and contains rare stem-like cells that are Let-7c negative or express both basal and luminal cytokeratins. ABM simulation of population dynamics of double cytokeratin cells supported increased self-renewal in irradiated MCF10A treated with TGFβ. Radiation-induced Notch concomitant with TGFβ was necessary for increased self-renewal of Let-7c negative MCF10A cells but not for EMT, indicating that these are independent processes. Consistent with these data, irradiating adult mice did not increase mammary repopulating activity or ER-negative tumors. These studies suggest that irradiation during puberty transiently increases stem cell self-renewal, which increases susceptibility to developing ER-negative breast cancer.
Collapse
Affiliation(s)
- Jonathan Tang
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Mazzarella L, Riva L, Luzi L, Ronchini C, Pelicci PG. The Genomic and Epigenomic Landscapes of AML. Semin Hematol 2014; 51:259-72. [DOI: 10.1053/j.seminhematol.2014.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
89
|
The Trp53-Trp53inp1-Tnfrsf10b pathway regulates the radiation response of mouse spermatogonial stem cells. Stem Cell Reports 2014; 3:676-89. [PMID: 25358794 PMCID: PMC4223695 DOI: 10.1016/j.stemcr.2014.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 08/05/2014] [Accepted: 08/06/2014] [Indexed: 12/22/2022] Open
Abstract
Germ cells are thought to exhibit a unique DNA damage response that differs from that of somatic stem cells, and previous studies suggested that Trp53 is not involved in the survival of spermatogonial stem cells (SSCs) after irradiation. Here, we report a critical role for the Trp53-Trp53inp1-Tnfrsf10b pathway during radiation-induced SSC apoptosis. Spermatogonial transplantation revealed that Trp53 deficiency increased the survival of SSCs after irradiation. Although Bbc3, a member of the intrinsic apoptotic pathway, was implicated in apoptosis of germ and somatic stem cells, Bbc3 depletion inhibited apoptosis in committed spermatogonia, but not in SSCs. In contrast, inhibition of Tnfrsf10b, an extrinsic apoptosis regulator, rescued SSCs. Tnfrsf10b, whose deficiency protected SSCs, was upregulated by Trp53inp1 upon irradiation. These results suggest that the Trp53-Trp53inp1-Tnfrsf10b pathway responds to genotoxic damage in SSCs and that stem and progenitor cells exhibit distinct DNA damage responses in self-renewing tissue. Trp53 induces radiation-induced apoptosis of spermatogonial stem cells (SSCs) Bbc3 induces radiation-induced apoptosis of spermatogonial progenitors Tnfsf10 is induced in spermatogonia and the SSC microenvironment Trp53inp1 upregulates Tnfrsf10b and induces SSC apoptosis upon irradiation
Collapse
|
90
|
Harburg G, Compton J, Liu W, Iwai N, Zada S, Marlow R, Strickland P, Zeng YA, Hinck L. SLIT/ROBO2 signaling promotes mammary stem cell senescence by inhibiting Wnt signaling. Stem Cell Reports 2014; 3:385-93. [PMID: 25241737 PMCID: PMC4266005 DOI: 10.1016/j.stemcr.2014.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 02/01/2023] Open
Abstract
WNT signaling stimulates the self-renewal of many types of adult stem cells, including mammary stem cells (MaSCs), but mechanisms that limit this activity are poorly understood. Here, we demonstrate that SLIT2 restricts stem cell renewal by signaling through ROBO2 in a subset of basal cells to negatively regulate WNT signaling. The absence of SLIT/ROBO2 signaling leads to increased levels of nuclear β-catenin. Robo2 loss does not increase the number of stem cells; instead, stem cell renewal is enhanced in the absence of SLIT/ROBO2 signaling. This is due to repressed expression of p16(INK4a), which, in turn, delays MaSC senescence. Together, our studies support a model in which SLITs restrict the expansion of MaSCs by countering the activity of WNTs and limiting self-renewal.
Collapse
Affiliation(s)
- Gwyndolen Harburg
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Jennifer Compton
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Wei Liu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Naomi Iwai
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Shahrzad Zada
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Rebecca Marlow
- Breakthrough Breast Cancer Unit, King's College London School of Medicine, London SE1 9RT, UK
| | - Phyllis Strickland
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Yi Arial Zeng
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lindsay Hinck
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA.
| |
Collapse
|
91
|
Pine SR, Liu W. Asymmetric cell division and template DNA co-segregation in cancer stem cells. Front Oncol 2014; 4:226. [PMID: 25191642 PMCID: PMC4139651 DOI: 10.3389/fonc.2014.00226] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/06/2014] [Indexed: 02/02/2023] Open
Abstract
During tissue homeostasis, normal stem cells self-renew and repopulate the diverse cell types found within the tissue via a series of carefully controlled symmetric and asymmetric cell divisions (ACDs). The notion that solid tumors comprise a subset of cancer stem cells (CSCs) with dysregulated self-renewal and excessive symmetric cell divisions has led to numerous studies aimed to elucidate the mechanisms regulating ACD under steady-state conditions, during stem-cell expansion and in cancer. In this perspective, we focus on a type of asymmetry that can be established during ACD, called non-random co-segregation of template DNA, which has been identified across numerous species, cell types, and cancers. We discuss the role of p53 loss in maintaining self-renewal in both normal and malignant cells. We then review our current knowledge of the mechanisms underlying co-segregation of template DNA strands and the stem-cell pathways associated with it in normal and CSCs.
Collapse
Affiliation(s)
- Sharon R Pine
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey , New Brunswick, NJ , USA
| | - Wenyu Liu
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey , New Brunswick, NJ , USA
| |
Collapse
|
92
|
Liu JC, Lerou PH, Lahav G. Stem cells: balancing resistance and sensitivity to DNA damage. Trends Cell Biol 2014; 24:268-74. [PMID: 24721782 PMCID: PMC4342985 DOI: 10.1016/j.tcb.2014.03.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/28/2014] [Accepted: 03/04/2014] [Indexed: 01/01/2023]
Abstract
Embryonic stem cells (ESCs) are known to be very sensitive to DNA damage and undergo rapid apoptosis even after low-damage doses. By contrast, adult stem cells show variable sensitivity to damage. Here we describe the multiple pathways that have been proposed to affect the sensitivity of stem cells to damage, including proximity to the apoptotic threshold (mitochondrial priming) and the p53 signaling pathway, through activation of transcription or direct interaction with proapoptotic proteins in the cytoplasm. We also discuss which cellular factors might connect mitochondrial priming with pluripotency and the potential therapeutic advances that can be achieved by better understanding of the molecular mechanisms leading to sensitivity or resistance of embryonic or adult stem cells from different tissues.
Collapse
Affiliation(s)
- Julia C Liu
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Paul H Lerou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Galit Lahav
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
93
|
McDermott N, Meunier A, Lynch TH, Hollywood D, Marignol L. Isogenic radiation resistant cell lines: development and validation strategies. Int J Radiat Biol 2014; 90:115-26. [PMID: 24350914 DOI: 10.3109/09553002.2014.873557] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE The comparison of cell lines with differing radiosensitivities and their molecular response to radiation exposure has been used in a number of human cancer models to study the molecular response to radiation. This review proposes to analyze and compare the protocols used by investigators for the development and validation of these isogenic models of radioresistance. CONCLUSION There is large variability in the strategies used to generate and validate isogenic models of radioresistance. Further characterization of these models is required.
Collapse
Affiliation(s)
- Niamh McDermott
- Radiation and Urologic Oncology, Applied Radiation Therapy Trinity and Prostate Molecular Oncology Research Group, Discipline of Radiation Therapy, Trinity College Dublin , Ireland
| | | | | | | | | |
Collapse
|
94
|
Yu J, Tu YK, Tang YB, Cheng NC. Stemness and transdifferentiation of adipose-derived stem cells using l-ascorbic acid 2-phosphate-induced cell sheet formation. Biomaterials 2014; 35:3516-26. [DOI: 10.1016/j.biomaterials.2014.01.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/08/2014] [Indexed: 12/29/2022]
|
95
|
Total body irradiation causes long-term mouse BM injury via induction of HSC premature senescence in an Ink4a- and Arf-independent manner. Blood 2014; 123:3105-15. [PMID: 24622326 DOI: 10.1182/blood-2013-07-515619] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Exposure to total body irradiation (TBI) induces not only acute hematopoietic radiation syndrome but also long-term or residual bone marrow (BM) injury. This residual BM injury is mainly attributed to permanent damage to hematopoietic stem cells (HSCs), including impaired self-renewal, decreased long-term repopulating capacity, and myeloid skewing. These HSC defects were associated with significant increases in production of reactive oxygen species (ROS), expression of p16(Ink4a) (p16) and Arf mRNA, and senescence-associated β-galacotosidase (SA-β-gal) activity, but not with telomere shortening or increased apoptosis, suggesting that TBI induces residual BM injury via induction of HSC premature senescence. This suggestion is supported by the finding that SA-β-gal(+) HSC-enriched LSK cells showed more pronounced defects in clonogenic activity in vitro and long-term engraftment after transplantation than SA-β-gal(-) LSK cells isolated from irradiated mice. However, genetic deletion of p16 and/or Arf had no effect on TBI-induced residual BM suppression and HSC senescence, because HSCs from irradiated p16 and/or Arf knockout (KO) mice exhibited changes similar to those seen in HSCs from wild-type mice after exposure to TBI. These findings provide important new insights into the mechanism by which TBI causes long-term BM suppression (eg, via induction of premature senescence of HSCs in a p16-Arf-independent manner).
Collapse
|
96
|
Behrens A, van Deursen JM, Rudolph KL, Schumacher B. Impact of genomic damage and ageing on stem cell function. Nat Cell Biol 2014; 16:201-7. [PMID: 24576896 PMCID: PMC4214082 DOI: 10.1038/ncb2928] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Impairment of stem cell function contributes to the progressive deterioration of tissue maintenance and repair with ageing. Evidence is mounting that age-dependent accumulation of DNA damage in both stem cells and cells that comprise the stem cell microenvironment are partly responsible for stem cell dysfunction with ageing. Here, we review the impact of the various types of DNA damage that accumulate with ageing on stem cell functionality, as well as the development of cancer. We discuss DNA-damage-induced cell intrinsic and extrinsic alterations that influence these processes, and review recent advances in understanding systemic adjustments to DNA damage and how they affect stem cells.
Collapse
Affiliation(s)
- Axel Behrens
- Mammalian Genetics Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, UK, and the School of Medicine, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine and the Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | - K Lenhard Rudolph
- Leibniz Institute of Age Research, Fritz Lipmann Institute e.V., Jena, 07745, Germany, and the Research Group on Molecular Aging, Faculty of Medicine, Friedrich-Schiller-University, Jena, Germany
| | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, 50931 Cologne, and the Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Institute for Genetics, and Systems Biology of Cologne, University of Cologne, Zülpicher Str. 47a, 50674 Cologne, Germany
| |
Collapse
|
97
|
Abstract
This review discusses the processes of DNA-damage-response and DNA-damage repair in stem and progenitor cells of several tissues. The long life-span of stem cells suggests that they may respond differently to DNA damage than their downstream progeny and, indeed, studies have begun to elucidate the unique stem cell response mechanisms to DNA damage. Because the DNA damage responses in stem cells and progenitor cells are distinctly different, stem and progenitor cells should be considered as two different entities from this point of view. Hematopoietic and mammary stem cells display a unique DNA-damage response, which involves active inhibition of apoptosis, entry into the cell-cycle, symmetric division, partial DNA repair and maintenance of self-renewal. Each of these biological events depends on the up-regulation of the cell-cycle inhibitor p21. Moreover, inhibition of apoptosis and symmetric stem cell division are the consequence of the down-regulation of the tumor suppressor p53, as a direct result of p21 up-regulation. A deeper understanding of these processes is required before these findings can be translated into human anti-aging and anti-cancer therapies. One needs to clarify and dissect the pathways that control p21 regulation in normal and cancer stem cells and define (a) how p21 blocks p53 functions in stem cells and (b) how p21 promotes DNA repair in stem cells. Is this effect dependent on p21s ability to inhibit p53? Such molecular knowledge may pave the way to methods for maintaining short-term tissue reconstitution while retaining long-term cellular and genomic integrity.
Collapse
|
98
|
Nakajima K, Crisma AR, Silva GB, Rogero MM, Fock RA, Borelli P. Malnutrition suppresses cell cycle progression of hematopoietic progenitor cells in mice via cyclin D1 down-regulation. Nutrition 2014; 30:82-9. [DOI: 10.1016/j.nut.2013.05.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 05/04/2013] [Accepted: 05/30/2013] [Indexed: 01/14/2023]
|
99
|
Abstract
Over the past few decades, advances in cancer research have enabled us to understand the different mechanisms that contribute to the aberrant proliferation of normal cells into abnormal cells that result in tumors. In the pursuit to find cures, researchers have primarily focused on various molecular level changes that are unique to cancerous cells. In humans, about 50 % or more cancers have a mutated tumor suppressor p53 gene thereby resulting in accumulation of p53 protein and losing its function to activate the target genes that regulate cell cycle and apoptosis. Extensive research conducted in murine cancer models with activated p53, loss of p53, or p53 missense mutations have facilitated researchers to understand the role of this key protein. Despite the identification of numerous triggers that causes lung cancer specific cure still remain elusive. One of the primary reasons attributed to this is due to the fact that the tumor tissue is heterogeneous and contains numerous sub-populations of cells. Studies have shown that a specific sub-population of cells termed as cancer stem cells (CSCs) drive the recurrence of cancer in response to standard chemotherapy. These CSCs are mutated cells with core properties similar to those of adult stem cells. They reside in a microenvironment within the tumor tissue that supports their growth and make them less susceptible to drug treatment. These cells possess properties of symmetric self-renewal and migration thus driving tumor formation and metastasis. Therefore, research specifically targeting these cells has gained prominence towards developing new therapeutic agents against cancer. This chapter focuses on lung cancer stem cells, p53 mutations noted in these cells, and importance of MDM2 interactions. Further, research approaches for better understanding of molecular mechanisms that drive CSC function and developing appropriate therapies are discussed.
Collapse
|
100
|
Abstract
Gadd45a has been involved in DNA damage response and in many malignancies, including leukemia. However, the function of Gadd45a in hematopoietic stem cells (HSCs) remains unknown. Here, we reported that Gadd45a-deficient (Gadd45a(-/-)) mice showed a normal hematologic phenotype under homeostatic conditions. However, following 5-fluorouracil treatment, Gadd45a(-/-) HSCs exhibited a faster recovery, associated with an increase in the proliferation rate. Interestingly, young Gadd45a(-/-) HSCs showed enhanced reconstitution ability in serial transplantation. Following ionizing radiation (IR), young Gadd45a(-/-) HSCs exhibited an increased resistance to IR-induced DNA damage, associated with a decrease in the apoptosis rate and delayed DNA repair. The significantly higher level of DNA damage in Gadd45a(-/-) HSCs ultimately promoted B-cell leukemia in further transplanted recipient mice. In old mice, Gadd45a(-/-) HSCs were functionally equal to wild-type HSCs but exhibited more DNA damage accumulation and increased sensitivity to IR than wild-type HSCs. In conclusion, Gadd45a plays a significant role in HSC stress responses. Gadd45a deficiency leads to DNA damage accumulation and impairment in apoptosis after exposure to IR, which increases the susceptibility of leukemogenesis.
Collapse
|