1
|
Valesyan S, Jora M, Addepalli B, Limbach PA. Stress-induced modification of Escherichia coli tRNA generates 5-methylcytidine in the variable loop. Proc Natl Acad Sci U S A 2024; 121:e2317857121. [PMID: 39495928 PMCID: PMC11572931 DOI: 10.1073/pnas.2317857121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 05/08/2024] [Indexed: 11/06/2024] Open
Abstract
There has been recent interest in trying to understand the connection between transfer RNA (tRNA) posttranscriptional modifications and changes in-cellular environmental conditions. Here, we report on the identification of the modified nucleoside 5-methylcytidine (m5C) in Escherichia coli tRNAs. This modification was determined to be present at position 49 of tRNA Tyr-QUA-II. Moreover, m5C levels in this tRNA are significantly elevated under high reactive oxygen specieis (ROS) conditions in E. coli cells. We identified the known ribosomal RNA methyltransferase rsmF as the enzyme responsible for m5C synthesis in tRNA and enzyme transcript levels are responsive to elevated levels of ROS in the cell. We further find that changes in m5C levels in this tRNA are not specific to Fenton-like reaction conditions elevating ROS, but heat shock can also induce increased modification of tRNA Tyr-QUA-II. Altogether, this work illustrates how cells adapt to changing environmental conditions through variations in tRNA modification profiles.
Collapse
Affiliation(s)
- Satenik Valesyan
- Department of Chemistry, Rieveschl Laboratories for Mass Spectrometry, University of Cincinnati, Cincinnati, OH45221-0172
| | - Manasses Jora
- Department of Chemistry, Rieveschl Laboratories for Mass Spectrometry, University of Cincinnati, Cincinnati, OH45221-0172
| | - Balasubrahmanyam Addepalli
- Department of Chemistry, Rieveschl Laboratories for Mass Spectrometry, University of Cincinnati, Cincinnati, OH45221-0172
| | - Patrick A. Limbach
- Department of Chemistry, Rieveschl Laboratories for Mass Spectrometry, University of Cincinnati, Cincinnati, OH45221-0172
| |
Collapse
|
2
|
Haines E, Nishida Y, Carr MI, Montoya RH, Ostermann LB, Zhang W, Zenke FT, Blaukat A, Andreeff M, Vassilev LT. DNA-PK inhibitor peposertib enhances p53-dependent cytotoxicity of DNA double-strand break inducing therapy in acute leukemia. Sci Rep 2021; 11:12148. [PMID: 34108527 PMCID: PMC8190296 DOI: 10.1038/s41598-021-90500-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
Peposertib (M3814) is a potent and selective DNA-PK inhibitor in early clinical development. It effectively blocks non-homologous end-joining repair of DNA double-strand breaks (DSB) and strongly potentiates the antitumor effect of ionizing radiation (IR) and topoisomerase II inhibitors. By suppressing DNA-PK catalytic activity in the presence of DNA DSB, M3814 potentiates ATM/p53 signaling leading to enhanced p53-dependent antitumor activity in tumor cells. Here, we investigated the therapeutic potential of M3814 in combination with DSB-inducing agents in leukemia cells and a patient-derived tumor. We show that in the presence of IR or topoisomerase II inhibitors, M3814 boosts the ATM/p53 response in acute leukemia cells leading to the elevation of p53 protein levels as well as its transcriptional activity. M3814 synergistically sensitized p53 wild-type, but not p53-deficient, AML cells to killing by DSB-inducing agents via p53-dependent apoptosis involving both intrinsic and extrinsic effector pathways. The antileukemic effect was further potentiated by enhancing daunorubicin-induced myeloid cell differentiation. Further, combined with the fixed-ratio liposomal formulation of daunorubicin and cytarabine, CPX-351, M3814 enhanced the efficacy against leukemia cells in vitro and in vivo without increasing hematopoietic toxicity, suggesting that DNA-PK inhibition could offer a novel clinical strategy for harnessing the anticancer potential of p53 in AML therapy.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Proliferation
- DNA Breaks, Double-Stranded
- DNA Repair
- DNA-Activated Protein Kinase/antagonists & inhibitors
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- Pyridazines/pharmacology
- Quinazolines/pharmacology
- Signal Transduction
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Eric Haines
- Translational Innovation Platform Oncology and Immuno-Oncology, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA
| | - Yuki Nishida
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael I Carr
- Translational Innovation Platform Oncology and Immuno-Oncology, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA
| | - Rafael Heinz Montoya
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren B Ostermann
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Weiguo Zhang
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frank T Zenke
- Translational Innovation Platform Oncology and Immuno-Oncology, Merck KGaA, Darmstadt, Germany
| | - Andree Blaukat
- Translational Innovation Platform Oncology and Immuno-Oncology, Merck KGaA, Darmstadt, Germany
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Lyubomir T Vassilev
- Translational Innovation Platform Oncology and Immuno-Oncology, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA.
| |
Collapse
|
3
|
Henry E, Arcangeli ML. How Hematopoietic Stem Cells Respond to Irradiation: Similarities and Differences between Low and High Doses of Ionizing Radiations. Exp Hematol 2020; 94:11-19. [PMID: 33290858 DOI: 10.1016/j.exphem.2020.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/12/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022]
Abstract
In this review, we will specifically address the newest insights on the effect of low doses of ionizing radiations on the hematopoietic stem cells, which are prone to long-term deleterious effects. Impact of high doses of irradiation on hematopoietic cells has been widely studied over the years, in line with the risk of accidental or terrorist exposure to irradiation and with a particular attention to the sensitivity of the hematopoietic system. Recently, more studies have focused on lower doses of irradiation on different tissues, due to the increasing exposure caused by medical imaging, radiotherapy or plane travelling for instance. Hence, we will delineate similarities and discrepancies in HSC response to high and low doses of irradiation from these studies.
Collapse
Affiliation(s)
- Elia Henry
- Team Niche and Cancer in Hematopoiesis, U1274, INSERM, 92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia/Service Stem Cells and Radiation/iRCM/JACOB/DRF, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université de Paris, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, CEA, Fontenay-aux-Roses, France
| | - Marie-Laure Arcangeli
- Team Niche and Cancer in Hematopoiesis, U1274, INSERM, 92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia/Service Stem Cells and Radiation/iRCM/JACOB/DRF, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université de Paris, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, CEA, Fontenay-aux-Roses, France.
| |
Collapse
|
4
|
Siberchicot C, Gault N, Déchamps N, Barroca V, Aguzzi A, Roméo PH, Radicella JP, Bravard A, Bernardino-Sgherri J. Prion protein deficiency impairs hematopoietic stem cell determination and sensitizes myeloid progenitors to irradiation. Haematologica 2019; 105:1216-1222. [PMID: 31371412 PMCID: PMC7193476 DOI: 10.3324/haematol.2018.205716] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Highly conserved among species and expressed in various types of cells, numerous roles have been attributed to the cellular prion protein (PrPC). In hematopoiesis, PrPC regulates hematopoietic stem cell self-renewal but the mechanisms involved in this regulation are unknown. Here we show that PrPC regulates hematopoietic stem cell number during aging and their determination towards myeloid progenitors. Furthermore, PrPC protects myeloid progenitors against the cytotoxic effects of total body irradiation. This radioprotective effect was associated with increased cellular prion mRNA level and with stimulation of the DNA repair activity of the Apurinic/pyrimidinic endonuclease 1, a key enzyme of the base excision repair pathway. Altogether, these results show a previously unappreciated role of PrPC in adult hematopoiesis, and indicate that PrPC-mediated stimulation of BER activity might protect hematopoietic progenitors from the cytotoxic effects of total body irradiation.
Collapse
Affiliation(s)
- Capucine Siberchicot
- French Alternative Energies and Atomic Energy Commission (CEA)/Direction of Fundamental Research (DRF)/Institute of Biology François Jacob (IBFJ)/Institute of Cellular and Molecular Radiobiology (iRCM), 92265 Fontenay-aux-Roses Cedex, France.,Laboratory of Research in Genetic Instability (LRIG), 92265 Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris-Sud, Paris, France
| | - Nathalie Gault
- French Alternative Energies and Atomic Energy Commission (CEA)/Direction of Fundamental Research (DRF)/Institute of Biology François Jacob (IBFJ)/Institute of Cellular and Molecular Radiobiology (iRCM), 92265 Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris-Sud, Paris, France.,Laboratory of Repair and Transcription in Hematopoietic Stem Cells (LRTS), 92265 Fontenay-aux-Roses Cedex, France.,Inserm U967, 92265 Fontenay-aux-Roses Cedex, France
| | - Nathalie Déchamps
- French Alternative Energies and Atomic Energy Commission (CEA)/Direction of Fundamental Research (DRF)/Institute of Biology François Jacob (IBFJ)/Institute of Cellular and Molecular Radiobiology (iRCM), 92265 Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris-Sud, Paris, France.,Inserm U967, 92265 Fontenay-aux-Roses Cedex, France
| | - Vilma Barroca
- French Alternative Energies and Atomic Energy Commission (CEA)/Direction of Fundamental Research (DRF)/Institute of Biology François Jacob (IBFJ)/Institute of Cellular and Molecular Radiobiology (iRCM), 92265 Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris-Sud, Paris, France.,Laboratory of Repair and Transcription in Hematopoietic Stem Cells (LRTS), 92265 Fontenay-aux-Roses Cedex, France.,Inserm U967, 92265 Fontenay-aux-Roses Cedex, France
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Paul-Henri Roméo
- French Alternative Energies and Atomic Energy Commission (CEA)/Direction of Fundamental Research (DRF)/Institute of Biology François Jacob (IBFJ)/Institute of Cellular and Molecular Radiobiology (iRCM), 92265 Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris-Sud, Paris, France.,Laboratory of Repair and Transcription in Hematopoietic Stem Cells (LRTS), 92265 Fontenay-aux-Roses Cedex, France.,Inserm U967, 92265 Fontenay-aux-Roses Cedex, France
| | - J Pablo Radicella
- French Alternative Energies and Atomic Energy Commission (CEA)/Direction of Fundamental Research (DRF)/Institute of Biology François Jacob (IBFJ)/Institute of Cellular and Molecular Radiobiology (iRCM), 92265 Fontenay-aux-Roses Cedex, France.,Laboratory of Research in Genetic Instability (LRIG), 92265 Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris-Sud, Paris, France
| | - Anne Bravard
- French Alternative Energies and Atomic Energy Commission (CEA)/Direction of Fundamental Research (DRF)/Institute of Biology François Jacob (IBFJ)/Institute of Cellular and Molecular Radiobiology (iRCM), 92265 Fontenay-aux-Roses Cedex, France .,Laboratory of Research in Genetic Instability (LRIG), 92265 Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris-Sud, Paris, France.,Laboratory of Repair and Transcription in Hematopoietic Stem Cells (LRTS), 92265 Fontenay-aux-Roses Cedex, France.,Inserm U967, 92265 Fontenay-aux-Roses Cedex, France
| | - Jacqueline Bernardino-Sgherri
- French Alternative Energies and Atomic Energy Commission (CEA)/Direction of Fundamental Research (DRF)/Institute of Biology François Jacob (IBFJ)/Institute of Cellular and Molecular Radiobiology (iRCM), 92265 Fontenay-aux-Roses Cedex, France .,Laboratory of Research in Genetic Instability (LRIG), 92265 Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Université Paris-Sud, Paris, France.,Laboratory of Repair and Transcription in Hematopoietic Stem Cells (LRTS), 92265 Fontenay-aux-Roses Cedex, France.,Inserm U967, 92265 Fontenay-aux-Roses Cedex, France
| |
Collapse
|
5
|
Parascandolo A, Laukkanen MO. Carcinogenesis and Reactive Oxygen Species Signaling: Interaction of the NADPH Oxidase NOX1-5 and Superoxide Dismutase 1-3 Signal Transduction Pathways. Antioxid Redox Signal 2019; 30:443-486. [PMID: 29478325 PMCID: PMC6393772 DOI: 10.1089/ars.2017.7268] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Reduction/oxidation (redox) balance could be defined as an even distribution of reduction and oxidation complementary processes and their reaction end products. There is a consensus that aberrant levels of reactive oxygen species (ROS), commonly observed in cancer, stimulate primary cell immortalization and progression of carcinogenesis. However, the mechanism how different ROS regulate redox balance is not completely understood. Recent Advances: In the current review, we have summarized the main signaling cascades inducing NADPH oxidase NOX1-5 and superoxide dismutase (SOD) 1-3 expression and their connection to cell proliferation, immortalization, transformation, and CD34+ cell differentiation in thyroid, colon, lung, breast, and hematological cancers. CRITICAL ISSUES Interestingly, many of the signaling pathways activating redox enzymes or mediating the effect of ROS are common, such as pathways initiated from G protein-coupled receptors and tyrosine kinase receptors involving protein kinase A, phospholipase C, calcium, and small GTPase signaling molecules. FUTURE DIRECTIONS The clarification of interaction of signal transduction pathways could explain how cells regulate redox balance and may even provide means to inhibit the accumulation of harmful levels of ROS in human pathologies.
Collapse
|
6
|
Anderson JJ. The relationship of mammal survivorship and body mass modeled by metabolic and vitality theories. POPUL ECOL 2018; 60:111-125. [PMID: 30546269 DOI: 10.1007/s10144-018-0617-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A model describes the relationship between mammal body mass and survivorship by combining replicative senescence theory postulating a cellular basis of aging, metabolic theory relating metabolism to body mass, and vitality theory relating survival to vitality loss and extrinsic mortality. In the combined framework, intrinsic mortality results from replicative senescence of the hematopoietic stem cells and extrinsic mortality results from environmental challenges. Because the model expresses the intrinsic and extrinsic rates with different powers of body mass, across the spectrum of mammals, survivorship changes from Type I to Type II curve shapes with decreasing body mass. Fitting the model to body mass and maximum lifespan data of 494 nonvolant mammals yields allometric relationships of body mass to the vitality parameters, from which full survivorship profiles were generated from body mass alone. Because maximum lifespan data is predominantly derived from captive populations, the generated survivorship curves were dominated by intrinsic mortality. Comparison of the mass-derived and observed survivorship curves provides insights into how specific populations deviate from the aggregate of populations observed under captivity.
Collapse
Affiliation(s)
- James J Anderson
- School of Aquatic and Fishery Sciences, University of Washington
| |
Collapse
|
7
|
Bruni E, Reichle A, Scimeca M, Bonanno E, Ghibelli L. Lowering Etoposide Doses Shifts Cell Demise From Caspase-Dependent to Differentiation and Caspase-3-Independent Apoptosis via DNA Damage Response, Inducing AML Culture Extinction. Front Pharmacol 2018; 9:1307. [PMID: 30483138 PMCID: PMC6243040 DOI: 10.3389/fphar.2018.01307] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/25/2018] [Indexed: 12/28/2022] Open
Abstract
Cytotoxic chemotherapy, still the most widely adopted anticancer treatment, aims at eliminating cancer cells inducing apoptosis with DNA damaging agents, exploiting the differential replication rate of cancer vs. normal cells; efficiency is evaluated in terms of extent of induced apoptosis, which depends on the individual cell sensitivity to a given drug, and on the dose. In this in vitro study, we report that the concentration of etoposide, a topoisomerase II poison widely used in clinics, determines both the kinetics of cell death, and the type of apoptosis induced. We observed that on a set of myeloid leukemia cell lines, etoposide at high (50 uM) dose promoted a rapid caspase-3-mediated apoptosis, whereas at low (0.5 uM) dose, it induced morphological and functional granulocytic differentiation and caspase-2-dependent, but caspase-3-independent, cell death, displaying features consistent with apoptosis. Both differentiation and caspase-2- (but not 3)-mediated apoptosis were contrasted by caffeine, a well-known inhibitor of the cellular DNA damage response (DDR), which maintained cell viability and cycling, indicating that the effects of low etoposide dose are not the immediate consequence of damage, but the result of a signaling pathway. DDR may be thus the mediator responsible for translating a mere dosage-effect into different signal transduction pathways, highlighting a strategic action in regulating timing and mode of cell death according to the severity of induced damage. The evidence of different molecular pathways induced by high vs. low drug doses may possibly contribute to explain the different effects of cytotoxic vs. metronomic therapy, the latter achieving durable clinical responses by treating cancer patients with stable, low doses of otherwise canonical cytotoxic drugs; intriguingly caspase-3, a major promoter of wounded tissue regeneration, is also a key factor of post-therapy cancer repopulation. All this suggests that cancer control in response to cytotoxic drugs arises from complex reprogramming mechanisms in tumor tissue, recently described as anakoinosis.
Collapse
Affiliation(s)
- Emanuele Bruni
- Department of Biology, University of Rome "Tor Vergata,", Rome, Italy
| | - Albrecht Reichle
- Department of Internal Medicine III, Haematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Manuel Scimeca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Elena Bonanno
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Diagnostica Medica and Villa dei Platani, Avellino, Italy
| | - Lina Ghibelli
- Department of Biology, University of Rome "Tor Vergata,", Rome, Italy
| |
Collapse
|
8
|
Naderi N, Najarkolaee SMG, Modanlookordi M, Shokrzadeh M, Irannejad H. Genoprotective Effect of New Triazine Derivatives in Endosulfan Mediated Toxicity, an In vivo and In vitro Study. LETT DRUG DES DISCOV 2018. [DOI: 10.2174/1570180815666180420095446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background:
Recently, we reported synthesis and neuroprotective activity of some new
1,2,4-triazine derivatives against H2O2 and β-amyloid toxicity in two neurotic cell lines, SHSY5Y
and PC12.
Methods:
The promising results obtained prompted us to further study on these potent neuroprotective
agents. In the current study, in vivo anti-inflammatory effect and also genoprotective activity of
these compounds in endosulfan-mediated toxicity were investigated. Compounds RT and SMO
exhibited high anti-inflammatory effect at 3 and 4 hours after injection in 20 mg/kg, and were even
more effective than Indomethacin (20 mg/kg).
Results:
Interestingly, compound SMO in 200 µM was the best compound in reducing micronuclei
significantly (P value <0.0001) in lymphocytes treated with endosulfan compared to control group.
Conclusion:
Herein, we report SMO as a genoprotective agent and a new drug candidate for endosulfan
mediated toxicity.
Collapse
Affiliation(s)
- Nima Naderi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mona Modanlookordi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Shokrzadeh
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Irannejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
9
|
D'Arena G, Seneca E, Migliaccio I, De Feo V, Giudice A, La Rocca F, Capunzo M, Calapai G, Festa A, Caraglia M, Musto P, Iorio EL, Ruggieri V. Oxidative stress in chronic lymphocytic leukemia: still a matter of debate. Leuk Lymphoma 2018; 60:867-875. [PMID: 30234409 DOI: 10.1080/10428194.2018.1509317] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is a large body of evidence showing a strong correlation between carcinogenesis of several types of human tumors, including chronic lymphocytic leukemia (CLL), and oxidative stress (OS). The mechanisms by which OS may promote cancer pathogenesis have not been completely deciphered yet and, in CLL, as in other neoplasms, whether OS is a primary cause or simply a downstream effect of the disease is still an open question. It has been demonstrated that, in CLL, OS concomitantly results from increased reactive oxygen species (ROS) production, mainly ascribable to CLL cells mitochondrial activity, and impaired antioxidant defenses. Interestingly, OS evaluation in CLL patients, at diagnosis, seems to have a prognostic significance, thus getting new insights in the biological comprehension of the disease with potential therapeutic implications.
Collapse
Affiliation(s)
- Giovanni D'Arena
- a Hematology and Stem Cell Transplantation Unit , IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture , Italy
| | - Elisa Seneca
- a Hematology and Stem Cell Transplantation Unit , IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture , Italy
| | - Ilaria Migliaccio
- a Hematology and Stem Cell Transplantation Unit , IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture , Italy
| | - Vincenzo De Feo
- b Pharmacology Department , University of Salerno , Salerno , Italy
| | - Aldo Giudice
- c Istituto Nazionale Tumori IRCCS Fondazione Pascale , Napoli , Italy
| | - Francesco La Rocca
- d Laboratory of Preclinical and Translational Research , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| | - Mario Capunzo
- e Department of Medicine and Surgery , University of Salerno , Salerno , Italy
| | - Gioacchino Calapai
- f Department of Biomedical and Dental Sciences and Morphological and Functional Sciences , University of Messina , Messina , Italy
| | - Agostino Festa
- g Department of Biochimics, Biophysics and General Pathology , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Michele Caraglia
- g Department of Biochimics, Biophysics and General Pathology , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Pellegrino Musto
- h Scientific Direction, IRCCS-CROB , Referral Cancer Center of Basilicata, Rionero in Vulture , Italy
| | | | - Vitalba Ruggieri
- d Laboratory of Preclinical and Translational Research , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| |
Collapse
|
10
|
Dhoke NR, Geesala R, Das A. Low Oxidative Stress-Mediated Proliferation Via JNK-FOXO3a-Catalase Signaling in Transplanted Adult Stem Cells Promotes Wound Tissue Regeneration. Antioxid Redox Signal 2018; 28:1047-1065. [PMID: 28826225 DOI: 10.1089/ars.2016.6974] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Aims: Stem cells exposed to pathological levels of reactive oxygen species (ROS) at wound sites fail to regenerate tissue. The molecular mechanism underlying differential levels of ROS-mediated regulation of stem cells remains elusive. This study elucidates the mechanistic role of catalase at 10 μM H2O2-induced proliferation of mouse bone marrow stromal (BMSC) and hematopoietic (HSPC) stem/progenitor cells. Results: BMSCs and HSPCs depicted an increased growth rate and colony formation, in the presence of 10 μM but not 100 μM concentration of H2O2, an effect that was perturbed by Vit. C. Mechanistically, JNK activation-FOXO3a nuclear translocation and binding of FOXO3a to catalase promoter at 10 μM H2O2 led to an increased expression and activity of anti-oxidant gene, catalase. This was followed by an increased proliferative phenotype via the AKT-dependent pathway that was perturbed in the presence of catalase-inhibitor, 3-aminotriazole due to an increased ROS-mediated inactivation of AKT. Preclinically, 10 μM H2O2-mediated preconditioning of BMSCs/HSPCs transplantation accelerated wound closure, enhanced catalase expression, and decreased ROS levels at the wound site. Transplantation of male donor cells into female recipient mice or GFP-labeled BMSCs or HSPCs depicted an increased engraftment and proliferation in preconditioned cell transplanted groups as compared with the wound control. Wound healing occurred via keratinocyte generation and vascularization in preconditioned BMSCs, whereas only neo-vascularization occurred in the preconditioned HSPCs transplanted groups. Innovation and Conclusion: Our study suggests a distinct role of catalase that protects BMSCs and HSPCs from low ROS and promotes proliferation. Transplantation of preconditioned stem cells enhanced wound tissue regeneration with a better antioxidant defense mechanism-as a therapeutic approach in stem cell transplantation-mediated tissue regeneration. Antioxid. Redox Signal. 28, 1047-1065.
Collapse
Affiliation(s)
- Neha R Dhoke
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Ramasatyaveni Geesala
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Amitava Das
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| |
Collapse
|
11
|
Senescent human hematopoietic progenitors show elevated expression of transposable elements and inflammatory genes. Exp Hematol 2018; 62:33-38.e6. [PMID: 29549053 DOI: 10.1016/j.exphem.2018.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 02/26/2018] [Accepted: 03/04/2018] [Indexed: 12/21/2022]
Abstract
Genomic transposable elements (TEs) constitute the majority of the genome. Expression of TEs is known to activate the double-stranded RNA recognition pathway ("viral mimicry"), leading to the activation of interferon-stimulated genes, inflammation, and immune-mediated cell death. Recently, we showed that the expression of TEs is suppressed along with immune pathways in leukemic stem cells (LSCs) in acute myeloid leukemia, suggesting a potential mechanism for immune escape of LSCs. This indicated that, during oncogenesis, where there is escape from senescence, expression of TEs is suppressed. Senescence is known to activate the interferon response and inflammatory cytokines, known as the senescence-associated secretory phenotype (SASP). We characterized the transcriptome of senescent and active human hematopoietic stem and progenitor cells (HSPCs) in vivo and showed co-occurrence of overexpression of TEs, SASP genes, and gene pathways of inflammation in senescence. The percentage of circulating senescent HSPCs (s-HSPCs) did not increase with age, indicating active clearance. Induction of senescence in human HSPCs in vitro showed increased expression of TE and SASP genes. SASP is known to mediate clearance of senescent cells and active clearance of senescent cells has been shown to increase organismal fitness. We speculate that the expression of TEs in s-HSPCs could contribute to orderly clearance of the cells via activation of immune pathways, warranting further mechanistic studies. This is the first study to characterize the transcriptome of human s-HSPCs in vivo, revealing activated expression of TEs and inflammatory genes.
Collapse
|
12
|
Cheng Y, Li Y, Ma C, Song Y, Xu H, Yu H, Xu S, Mu Q, Li H, Chen Y, Zhao G. Arsenic trioxide inhibits glioma cell growth through induction of telomerase displacement and telomere dysfunction. Oncotarget 2017; 7:12682-92. [PMID: 26871293 PMCID: PMC4914314 DOI: 10.18632/oncotarget.7259] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/24/2016] [Indexed: 12/22/2022] Open
Abstract
Glioblastomas are resistant to many kinds of treatment, including chemotherapy, radiation and other adjuvant therapies. As2O3 reportedly induces ROS generation in cells, suggesting it may be able to induce telomerase suppression and telomere dysfunction in glioblastoma cells. We show here that As2O3 induces ROS generation as well as telomerase phosphorylation in U87, U251, SHG4 and C6 glioma cells. It also induces translocation of telomerase from the nucleus to the cytoplasm, thereby decreasing total telomerase activity. These effects of As2O3 trigger an extensive DNA damage response at the telomere, which includes up-regulation of ATM, ATR, 53BP1, γ-H2AX and Mer11, in parallel with telomere fusion and 3′-overhang degradation. This ultimately results in induction of p53- and p21-mediated cell apoptosis, G2/M cell cycle arrest and cellular senescence. These results provide new insight into the antitumor effects of As2O3 and can perhaps contribute to solving the problem of glioblastoma treatment resistance.
Collapse
Affiliation(s)
- Ye Cheng
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Yunqian Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Chengyuan Ma
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Yang Song
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Haiyang Xu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Hongquan Yu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Songbai Xu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Qingchun Mu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Haisong Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Yong Chen
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Gang Zhao
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| |
Collapse
|
13
|
Shubernetskaya O, Skvortsov D, Evfratov S, Rubtsova M, Belova E, Strelkova O, Cherepaninets V, Zhironkina O, Olovnikov A, Zvereva M, Dontsova O, Kireev I. Interstitial telomeric repeats-associated DNA breaks. Nucleus 2017; 8:641-653. [PMID: 28914588 PMCID: PMC5788545 DOI: 10.1080/19491034.2017.1356501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 06/28/2017] [Accepted: 07/04/2017] [Indexed: 02/08/2023] Open
Abstract
During a cell's lifespan, DNA break formation is a common event, associated with many processes, from replication to apoptosis. Most of DNA breaks are readily repaired, but some are meant to persist in time, such as the chromosome ends, protected by telomeres. Besides them, eukaryotic genomes comprise shorter stretches of interstitial telomeric repeats. We assumed that the latter may also be associated with the formation of DNA breaks meant to persist in time. In zebrafish and mouse embryos, cells containing numerous breakage foci were identified. These breaks were not associated with apoptosis or replication, nor did they seem to activate DNA damage response machinery. Unlike short-living, accidental sparse breaks, the ones we found seem to be closely associated, forming discrete break foci. A PCR-based method was developed, allowing specific amplification of DNA regions located between inverted telomeric repeats associated with breaks. The cloning and sequencing of such DNA fragments were found to denote some specificity in their distribution for different tissue types and development stages.
Collapse
Affiliation(s)
- Olga Shubernetskaya
- Chemistry Department, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry Skvortsov
- Chemistry Department, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Sergey Evfratov
- Chemistry Department, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Maria Rubtsova
- Chemistry Department, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Elena Belova
- Chemistry Department, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Olga Strelkova
- A.N. Belozersky Institute of Physico-chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Varvara Cherepaninets
- A.N. Belozersky Institute of Physico-chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Oxana Zhironkina
- A.N. Belozersky Institute of Physico-chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | | | - Maria Zvereva
- Chemistry Department, M.V. Lomonosov Moscow State University, Moscow, Russia
- A.N. Belozersky Institute of Physico-chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Olga Dontsova
- Chemistry Department, M.V. Lomonosov Moscow State University, Moscow, Russia
- A.N. Belozersky Institute of Physico-chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia
| | - Igor Kireev
- A.N. Belozersky Institute of Physico-chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
14
|
Wu L, Cole A, Du W. Immune-DDR crosstalk in pre-leukemia stem cells. Oncotarget 2017; 8:81731-81732. [PMID: 29137216 PMCID: PMC5669842 DOI: 10.18632/oncotarget.21047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Indexed: 11/25/2022] Open
Affiliation(s)
- Limei Wu
- Wei Du: Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Allison Cole
- Wei Du: Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Wei Du
- Wei Du: Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
15
|
The pan-Bcl2 Inhibitor AT101 Activates the Intrinsic Apoptotic Pathway and Causes DNA Damage in Acute Myeloid Leukemia Stem-Like Cells. Target Oncol 2017; 12:677-687. [DOI: 10.1007/s11523-017-0509-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
16
|
Delia D, Mizutani S. The DNA damage response pathway in normal hematopoiesis and malignancies. Int J Hematol 2017; 106:328-334. [PMID: 28707218 DOI: 10.1007/s12185-017-2300-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/05/2017] [Indexed: 11/29/2022]
Abstract
In mammalian cells, the DNA damage response (DDR) prevents the replication and propagation of DNA errors to the next generation, thus maintaining genomic stability. At the heart of the DDR are the related signaling kinases ATM, ATR, and DNA-PK, which regulate DNA repair and associated events such as cell cycle checkpoints, chromatin remodeling, transcription, and ultimately apoptosis. Several findings highlight the occurrence of DDR in hemopoietic stem cells (HSCs), and persistence of DNA lesions in these cells promotes their functional decline and accumulation of leukemogenic mutations. Besides favoring tumor formation and progression, molecular defects that directly or indirectly inactivate certain DDR pathways can provide a therapeutic opportunity, since a reduced ability to repair DNA lesions renders hemopoietic malignancies vulnerable to genotoxic drugs acting also through synthetic lethal interactions. Here, we discuss the essential role of DDR in HSC maintenance and protection against leukemogenesis, and how acquired DDR dysfunctions or pharmacological agents that block this pathway can be effectively exploited for the treatment of various hematopoietic malignancies.
Collapse
Affiliation(s)
- Domenico Delia
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| | - Shuki Mizutani
- Kawasaki North Center for Childhood Developmental Disorder/Tokyo Medical and Dental University, 5-26-1 Katahira, Aso-ku, Kawasaki, 215-0003, Japan
| |
Collapse
|
17
|
De Nicola M, Bruni E, Traversa E, Ghibelli L. Slow release of etoposide from dextran conjugation shifts etoposide activity from cytotoxicity to differentiation: A promising tool for dosage control in anticancer metronomic therapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2005-2014. [PMID: 28535989 DOI: 10.1016/j.nano.2017.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/30/2017] [Accepted: 05/08/2017] [Indexed: 11/27/2022]
Abstract
Drug conjugation, improving drug stability, solubility and body permanence, allows achieving impressive results in tumor control. Here, we show that conjugation may provide a straightforward method to administer drugs by the emerging anticancer metronomic approach, presently consisting of low, repeated doses of cytotoxic drugs used in traditional chemotherapy, thus reducing toxicity without reducing efficiency; however, low dose maintenance in tumor sites is difficult. We show that conjugating the antitumor drug etoposide to dextran via pH-sensitive bond produces slow releasing, apoptosis-proficient conjugates rapidly internalized into acidic lysosomes; importantly, release of active etoposide requires cell internalization and acidic pH. Conjugation, without impairing etoposide-induced complete elimination of tumor cells, shifted the mode of apoptosis from cytotoxicity- to differentiation-related; interestingly, high conjugate doses acted as low doses of free etoposide, thus mimicking the effect of metronomic therapy. This indicates slow release as a promising novel strategy for stabilizing low drug levels in metronomic regimens.
Collapse
Affiliation(s)
- Milena De Nicola
- Dipartimento di Biologia, Università di Roma Tor Vergata, Roma, Italy; Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Roma, Italy.
| | - Emanuele Bruni
- Dipartimento di Biologia, Università di Roma Tor Vergata, Roma, Italy.
| | - Enrico Traversa
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Roma, Italy; International Research Center for Renewable Energy (IRCRE), Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Lina Ghibelli
- Dipartimento di Biologia, Università di Roma Tor Vergata, Roma, Italy.
| |
Collapse
|
18
|
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
|
19
|
The immune receptor Trem1 cooperates with diminished DNA damage response to induce preleukemic stem cell expansion. Leukemia 2016; 31:423-433. [PMID: 27568523 DOI: 10.1038/leu.2016.242] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 07/26/2016] [Accepted: 08/09/2016] [Indexed: 12/30/2022]
Abstract
Fanconi anemia (FA) is an inherited bone marrow failure syndrome with extremely high risk of leukemic transformation. Here we investigate the relationship between DNA damage response (DDR) and leukemogenesis using the Fanca knockout mouse model. We found that chronic exposure of the Fanca-/- hematopoietic stem cells to DNA crosslinking agent mitomycin C in vivo leads to diminished DDR, and the emergence/expansion of pre-leukemia stem cells (pre-LSCs). Surprisingly, although genetic correction of Fanca deficiency in the pre-LSCs restores DDR and reduces genomic instability, but fails to prevent pre-LSC expansion or delay leukemia development in irradiated recipients. Furthermore, we identified transcription program underlying dysregulated DDR and cell migration, myeloid proliferation, and immune response in the Fanca-/- pre-LSCs. Forced expression of the downregulated DNA repair genes, Rad51c or Trp53i13, in the Fanca-/- pre-LSCs partially rescues DDR but has no effect on leukemia, whereas shRNA knockdown of the upregulated immune receptor genes Trem1 or Pilrb improves leukemia-related survival, but not DDR or genomic instability. Furthermore, Trem1 cooperates with diminished DDR in vivo to promote Fanca-/- pre-LSC expansion and leukemia development. Our study implicates diminishing DDR as a root cause of FA leukemogenesis, which subsequently collaborates with other signaling pathways for leukemogenic transformation.
Collapse
|
20
|
Mujoo K, Butler EB, Pandita RK, Hunt CR, Pandita TK. Pluripotent Stem Cells and DNA Damage Response to Ionizing Radiations. Radiat Res 2016; 186:17-26. [PMID: 27332952 PMCID: PMC4963261 DOI: 10.1667/rr14417.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pluripotent stem cells (PSCs) hold great promise in regenerative medicine, disease modeling, functional genomics, toxicological studies and cell-based therapeutics due to their unique characteristics of self-renewal and pluripotency. Novel methods for generation of pluripotent stem cells and their differentiation to the specialized cell types such as neuronal cells, myocardial cells, hepatocytes and beta cells of the pancreas and many other cells of the body are constantly being refined. Pluripotent stem cell derived differentiated cells, including neuronal cells or cardiac cells, are ideal for stem cell transplantation as autologous or allogeneic cells from healthy donors due to their minimal risk of rejection. Radiation-induced DNA damage, ultraviolet light, genotoxic stress and other intrinsic and extrinsic factors triggers a series of biochemical reactions known as DNA damage response. To maintain genomic stability and avoid transmission of mutations into progenitors cells, stem cells have robust DNA damage response signaling, a contrast to somatic cells. Stem cell transplantation may protect against radiation-induced late effects. In particular, this review focuses on differential DNA damage response between stem cells and derived differentiated cells and the possible pathways that determine such differences.
Collapse
Affiliation(s)
- Kalpana Mujoo
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| | - E. Brian Butler
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| | - Raj K. Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| | - Clayton R. Hunt
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| | - Tej K. Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| |
Collapse
|
21
|
Wingert S, Rieger MA. Terminal differentiation induction as DNA damage response in hematopoietic stem cells by GADD45A. Exp Hematol 2016; 44:561-6. [PMID: 27262218 DOI: 10.1016/j.exphem.2016.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 12/17/2022]
Abstract
Hematopoietic stem cells (HSCs) sustain lifelong blood cell regeneration by balancing their ability for self-renewal with their ability to differentiate into all blood cell types. To prevent organ exhaustion and malignant transformation, long-lived HSCs, in particular, must be protected from exogenous and endogenous stress, which cause severe DNA damage. When DNA is damaged, distinct DNA repair mechanisms and cell fate controls occur in adult HSCs compared with committed cells. Growth arrest and DNA damage-inducible 45 alpha (GADD45A) is known to coordinate a variety of cellular stress responses, indicating the molecule is an important stress mediator. So far, the function of GADD45A in hematopoietic stem and progenitor cells is controversial and appears highly dependent on the cell type and stress stimulus. Recent studies have analyzed its role in cell fate decision control of prospectively isolated HSCs and have revealed unexpected functions of GADD45A, as discussed here. The upregulation of GADD45A by DNA damage-causing conditions results in enhanced HSC differentiation, probably to efficiently eliminate aberrant HSCs from the system. These findings, in concert with a few studies on other stem cell systems, have led us to propose DNA damage-induced differentiation as a novel DNA damage response mechanism in stem cells that circumvents the fatal consequences of cumulative DNA damage in the stem cell compartment.
Collapse
Affiliation(s)
- Susanne Wingert
- LOEWE Center for Cell and Gene Therapy and Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Michael A Rieger
- LOEWE Center for Cell and Gene Therapy and Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
22
|
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
|
23
|
Hao S, Chen C, Cheng T. Cell cycle regulation of hematopoietic stem or progenitor cells. Int J Hematol 2016; 103:487-97. [DOI: 10.1007/s12185-016-1984-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 11/24/2022]
|
24
|
Solhaug A, Wisbech C, Christoffersen T, Hult L, Lea T, Eriksen G, Holme J. The mycotoxin alternariol induces DNA damage and modify macrophage phenotype and inflammatory responses. Toxicol Lett 2015; 239:9-21. [DOI: 10.1016/j.toxlet.2015.08.1107] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/24/2015] [Accepted: 08/28/2015] [Indexed: 02/07/2023]
|