1
|
Ni Y, Chen H, Zhan Q, Zhuang Z. Nuclear export of PML promotes p53-mediated apoptosis and ferroptosis. Cell Signal 2024; 121:111278. [PMID: 38944257 DOI: 10.1016/j.cellsig.2024.111278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Promyelocytic leukemia protein (PML), a tumor suppressor protein, plays a key role in cell cycle regulation, apoptosis, senescence and cellular metabolism. Here, we report that PML promotes apoptosis and ferroptosis. Our data showed that PML over-expression inhibited cell proliferation and migration. PML over-expression increased apoptotic cells, nuclear condensation and the loss of mitochondrial membrane potential, accompanied by regulation of Bcl-2 family proteins and reactive oxygen species (ROS) level, suggesting that PML enhanced apoptosis. Meanwhile, PML over-expression not only increased lipid ROS accumulation and Malondialdehyde (MDA) content but also downregulated solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) expression, indicating that PML enhanced ferroptosis. Additionally, knockdown of p53 attenuated the effect of PML on SLC7A11 and GPX4, and inhibited the increase of lipid ROS and ROS by PML over-expression. Moreover, translocation of PML from nucleus to cytoplasm not only promoted apoptosis and ferroptosis, but also inhibited cell proliferation. Taken together, PML promotes apoptosis and ferroptosis, in which the mediation of p53 and the nuclear export of PML play important roles.
Collapse
Affiliation(s)
- Yue Ni
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; Centre for Optical and Electromagnetic Research, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510631, China
| | - Hongce Chen
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Qiuqiang Zhan
- Centre for Optical and Electromagnetic Research, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510631, China
| | - Zhengfei Zhuang
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
2
|
Tang L, Zhu Y, Du Y, Long X, Long Y, Tang Y, Liu J. Clinicopathologic features and genomic profiling of female axillary lymph node metastases from adenocarcinoma or poorly differentiated carcinoma of unknown primary. J Cancer Res Clin Oncol 2024; 150:256. [PMID: 38750402 PMCID: PMC11096249 DOI: 10.1007/s00432-024-05783-6] [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: 03/16/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024]
Abstract
PURPOSE Axillary lymph node metastases from adenocarcinoma or poorly differentiated carcinoma of unknown primary (CUPAx) is a rare disease in women. This retrospective study intended to examine the clinicopathological features of CUPAx and compared CUPAx genetically with axillary lymph node metastases from breast cancer (BCAx), investigating differences in their biological behavior. METHODS We conducted the clinical and prognostic analysis of 58 CUPAx patients in West China Hospital spanning from 2009 to 2021. Gemonic profiling of 12 CUPAx patients and 16 BCAx patients was conducted by the FoundationOne CDx (F1CDx) platform. Moreover, we also compared the gene mutation spectrum and relevant pathways between the two groups and both TCGA and COSMIC databases. RESULTS The majority of the 58 CUPAx patients were HR-/HER2- subtype. Most patients received mastectomy combined radiotherapy (50 Gy/25f). CUPAx patients who received mastectomy instead of breast-conserving surgery had a more favorable overall prognosis. Radiotherapy in chest wall/breast and supraclavicular/infraclavicular fossa was the independent prognostic factor (HR = 0.05, 95%CI = 0.00-0.93, P = 0.04). In 28 sequencing samples (CUPAx, n = 12, BCAx, n = 16) and 401 TCGA-BRCA patients, IRS2 only mutated in CUPAx (33.33%) but amplified in BCAx (11.11%) and TCGA-BRCA (1.5%). Pathway analysis revealed that BCAx had more NOTCH pathway mutations than CUPAx. Enrichment analysis showed that CUPAx enriched more in mammary development and PML bodies than BCAx, but less in the positive regulation of kinase activity. CONCLUSIONS More active treatment methods, like chemotherapy, mastectomy and postoperative radiotherapy, could improve the prognosis of CUPAx. The differential mutation genes of CUPAx and BCAx might be associated with their respective biological behaviors like invasiveness and prognosis.
Collapse
Affiliation(s)
- Liansha Tang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
- Biotherapy Clinical Research Center of Sichuan Province, Chengdu, 610041, China
| | - Yueting Zhu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
- Biotherapy Clinical Research Center of Sichuan Province, Chengdu, 610041, China
| | - Yang Du
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
- Biotherapy Clinical Research Center of Sichuan Province, Chengdu, 610041, China
| | - Xiangyu Long
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
- Biotherapy Clinical Research Center of Sichuan Province, Chengdu, 610041, China
| | - Yixiu Long
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China
| | - Yuan Tang
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Jiyan Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China.
- Biotherapy Clinical Research Center of Sichuan Province, Chengdu, 610041, China.
| |
Collapse
|
3
|
Bercier P, de Thé H. History of Developing Acute Promyelocytic Leukemia Treatment and Role of Promyelocytic Leukemia Bodies. Cancers (Basel) 2024; 16:1351. [PMID: 38611029 PMCID: PMC11011038 DOI: 10.3390/cancers16071351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
The story of acute promyelocytic leukemia (APL) discovery, physiopathology, and treatment is a unique journey, transforming the most aggressive form of leukemia to the most curable. It followed an empirical route fueled by clinical breakthroughs driving major advances in biochemistry and cell biology, including the discovery of PML nuclear bodies (PML NBs) and their central role in APL physiopathology. Beyond APL, PML NBs have emerged as key players in a wide variety of biological functions, including tumor-suppression and SUMO-initiated protein degradation, underscoring their broad importance. The APL story is an example of how clinical observations led to the incremental development of the first targeted leukemia therapy. The understanding of APL pathogenesis and the basis for cure now opens new insights in the treatment of other diseases, especially other acute myeloid leukemias.
Collapse
Affiliation(s)
- Pierre Bercier
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75231 Paris, France;
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, 75010 Paris, France
| | - Hugues de Thé
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75231 Paris, France;
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, 75010 Paris, France
- Hematology Laboratory, Hôpital St Louis, AP/HP, 75010 Paris, France
| |
Collapse
|
4
|
Martinez TC, McNerney ME. Haploinsufficient Transcription Factors in Myeloid Neoplasms. ANNUAL REVIEW OF PATHOLOGY 2024; 19:571-598. [PMID: 37906947 DOI: 10.1146/annurev-pathmechdis-051222-013421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Many transcription factors (TFs) function as tumor suppressor genes with heterozygous phenotypes, yet haploinsufficiency generally has an underappreciated role in neoplasia. This is no less true in myeloid cells, which are normally regulated by a delicately balanced and interconnected transcriptional network. Detailed understanding of TF dose in this circuitry sheds light on the leukemic transcriptome. In this review, we discuss the emerging features of haploinsufficient transcription factors (HITFs). We posit that: (a) monoallelic and biallelic losses can have distinct cellular outcomes; (b) the activity of a TF exists in a greater range than the traditional Mendelian genetic doses; and (c) how a TF is deleted or mutated impacts the cellular phenotype. The net effect of a HITF is a myeloid differentiation block and increased intercellular heterogeneity in the course of myeloid neoplasia.
Collapse
Affiliation(s)
- Tanner C Martinez
- Department of Pathology, Department of Pediatrics, Section of Hematology/Oncology, The University of Chicago Medicine Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois, USA;
- Medical Scientist Training Program, The University of Chicago, Chicago, Illinois, USA
| | - Megan E McNerney
- Department of Pathology, Department of Pediatrics, Section of Hematology/Oncology, The University of Chicago Medicine Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois, USA;
| |
Collapse
|
5
|
Rérolle D, de Thé H. The PML hub: An emerging actor of leukemia therapies. J Exp Med 2023; 220:e20221213. [PMID: 37382966 PMCID: PMC10309189 DOI: 10.1084/jem.20221213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/29/2023] [Accepted: 06/09/2023] [Indexed: 06/30/2023] Open
Abstract
PML assembles into nuclear domains that have attracted considerable attention from cell and cancer biologists. Upon stress, PML nuclear bodies modulate sumoylation and other post-translational modifications, providing an integrated molecular framework for the multiple roles of PML in apoptosis, senescence, or metabolism. PML is both a sensor and an effector of oxidative stress. Emerging data has demonstrated its key role in promoting therapy response in several hematological malignancies. While these membrane-less nuclear hubs can enforce efficient cancer cell clearance, their downstream pathways deserve better characterization. PML NBs are druggable and their known modulators may have broader clinical utilities than initially thought.
Collapse
Affiliation(s)
- Domitille Rérolle
- Center for Interdisciplinary Research in Biology, Collège de France, Inserm, PSL Research University, Paris, France
- Université Paris Cité, Inserm U944, CNRS, GenCellDis, Institut de Recherche Saint-Louis, Paris, France
| | - Hugues de Thé
- Center for Interdisciplinary Research in Biology, Collège de France, Inserm, PSL Research University, Paris, France
- Université Paris Cité, Inserm U944, CNRS, GenCellDis, Institut de Recherche Saint-Louis, Paris, France
- Chaire d'Oncologie Cellulaire et Moléculaire, Collège de France, Paris, France
- Service d'Hématologie Biologique, Assistance Publique-Hôpitaux de Paris, Hôpital St. Louis, Paris, France
| |
Collapse
|
6
|
Garufi A, D’Orazi V, Pistritto G, Cirone M, D’Orazi G. The Sweet Side of HIPK2. Cancers (Basel) 2023; 15:2678. [PMID: 37345014 PMCID: PMC10216817 DOI: 10.3390/cancers15102678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
HIPK2 is an evolutionary conserved protein kinase which modulates many molecular pathways involved in cellular functions such as apoptosis, DNA damage response, protein stability, and protein transcription. HIPK2 plays a key role in the cancer cell response to cytotoxic drugs as its deregulation impairs drug-induced cancer cell death. HIPK2 has also been involved in regulating fibrosis, angiogenesis, and neurological diseases. Recently, hyperglycemia was found to positively and/or negatively regulate HIPK2 activity, affecting not only cancer cell response to chemotherapy but also the progression of some diabetes complications. The present review will discuss how HIPK2 may be influenced by the high glucose (HG) metabolic condition and the consequences of such regulation in medical conditions.
Collapse
Affiliation(s)
- Alessia Garufi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Valerio D’Orazi
- Department of Surgery, Sapienza University, 00185 Rome, Italy;
| | - Giuseppa Pistritto
- Centralized Procedures Office, Italian Medicines Agency (AIFA), 00187 Rome, Italy;
| | - Mara Cirone
- Laboratory Affiliated to Pasteur Institute Italy Foundation Cenci Bolognetti, Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Gabriella D’Orazi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy
| |
Collapse
|
7
|
Shi X, Du Y, Li S, Wu H. The Role of SUMO E3 Ligases in Signaling Pathway of Cancer Cells. Int J Mol Sci 2022; 23:3639. [PMID: 35408996 PMCID: PMC8998487 DOI: 10.3390/ijms23073639] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Small ubiquitin-like modifier (SUMO)ylation is a reversible post-translational modification that plays a crucial role in numerous aspects of cell physiology, including cell cycle regulation, DNA damage repair, and protein trafficking and turnover, which are of importance for cell homeostasis. Mechanistically, SUMOylation is a sequential multi-enzymatic process where SUMO E3 ligases recruit substrates and accelerate the transfer of SUMO onto targets, modulating their interactions, localization, activity, or stability. Accumulating evidence highlights the critical role of dysregulated SUMO E3 ligases in processes associated with the occurrence and development of cancers. In the present review, we summarize the SUMO E3 ligases, in particular, the novel ones recently identified, and discuss their regulatory roles in cancer pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Huijian Wu
- School of Bioengineering & Province Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian 116024, China; (X.S.); (Y.D.); (S.L.)
| |
Collapse
|
8
|
Michaille JJ, Awad H, Fortman EC, Efanov AA, Tili E. miR-155 expression in antitumor immunity: The higher the better? Genes Chromosomes Cancer 2019; 58:208-218. [PMID: 30382602 DOI: 10.1002/gcc.22698] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/25/2018] [Accepted: 10/28/2018] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs are small noncoding RNAs that modulate gene expression either directly, by impairing the stability and/or translation of transcripts that contain their specific target sequence, or indirectly through the targeting of transcripts that encode transcription factors, factors implicated in signal transduction pathways, or epigenetic regulators. Abnormal expression of micro-RNAs has been found in nearly all types of pathologies, including cancers. MiR-155 has been the first microRNA to be implicated in the regulation of the innate and adaptative immune responses, and its expression is either increased or decreased in a variety of liquid and solid malignancies. In this review, we examine the oncogenic and antitumor potentials of miR-155, with special emphasize on its dose-dependent effects. We describe the impact of miR-155 levels on antitumor activity of lymphocytes and myeloid cells. We discuss miR-155 dose-dependent effects in leukemias and analyze results showing that miR-155 intermediate levels tend to be detrimental, whereas high levels of miR-155 expression usually prove beneficial. We also examine the beneficial effects of high levels of miR-155 expression in solid tumors. We discuss the possible causal involvement of miR-155 in leukemias and dementia in individuals with Down's syndrome. We finally propose that increasing miR-155 levels in immune cells might increase the efficiency of newly developed cancer immunotherapies, due to miR-155 ability to target transcripts encoding immune checkpoints such as cytotoxic T lymphocyte antigen-4 or programmed death-ligand 1.
Collapse
Affiliation(s)
- Jean-Jacques Michaille
- BioPerox-IL, Université de Bourgogne-Franche Comté (EA 7270), Dijon, France.,Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Hamdy Awad
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Emily C Fortman
- Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Alexander A Efanov
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Esmerina Tili
- Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, Ohio.,Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
9
|
Narayan N, Bracken CP, Ekert PG. MicroRNA-155 expression and function in AML: An evolving paradigm. Exp Hematol 2018; 62:1-6. [PMID: 29601851 DOI: 10.1016/j.exphem.2018.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/22/2018] [Accepted: 03/23/2018] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) arises when immature myeloid blast cells acquire multiple, recurrent genetic and epigenetic changes that result in dysregulated proliferation. Acute leukemia is the most common form of pediatric cancer, with AML accounting for ~20% of all leukemias in children. The genomic aberrations that drive AML inhibit myeloid differentiation and activate signal transduction pathways that drive proliferation. MicroRNAs, a class of small (~22 nucleotide) noncoding RNAs that posttranscriptionally suppress the expression of specifically targeted transcripts, are also frequently dysregulated in AML, which may prove useful for the purposes of disease classification, prognosis, and future therapeutic approaches. MicroRNA expression profiles are associated with patient prognosis and responses to standard chemotherapy, including predicting therapy resistance in AML. miR-155 is the primary focus of this review because it has been repeatedly associated with poorer survival across multiple cohorts of adult and pediatric AML. We discuss some novel features of miR-155 expression in AML, in particular how the levels of expression can critically influence function. Understanding the role of microRNAs in AML and the ways in which microRNA expression influences AML biology is one means to develop novel and more targeted therapies.
Collapse
Affiliation(s)
- Nisha Narayan
- Murdoch Childrens Research Institute, Parkville, 3052, Australia
| | - Cameron P Bracken
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5000, Australia
| | - Paul G Ekert
- Murdoch Childrens Research Institute, Parkville, 3052, Australia.
| |
Collapse
|
10
|
Barrdahl M, Canzian F, Gaudet MM, Gapstur SM, Trichopoulou A, Tsilidis K, van Gils CH, Borgquist S, Weiderpass E, Khaw KT, Giles GG, Milne RL, Le Marchand L, Haiman C, Lindström S, Kraft P, Hunter DJ, Ziegler R, Chanock SJ, Yang XR, Buring JE, Lee IM, Kaaks R, Campa D. A comprehensive analysis of polymorphic variants in steroid hormone and insulin-like growth factor-1 metabolism and risk of in situ breast cancer: Results from the Breast and Prostate Cancer Cohort Consortium. Int J Cancer 2018; 142:1182-1188. [PMID: 29114882 DOI: 10.1002/ijc.31145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/26/2017] [Accepted: 08/04/2017] [Indexed: 11/08/2022]
Abstract
We assessed the association between 1,414 single nucleotide polymorphisms (SNPs) in genes involved in synthesis and metabolism of steroid hormones and insulin-like growth factor 1, and risk of breast cancer in situ (BCIS), with the aim of determining whether any of these were disease specific. This was carried out using 1,062 BCIS cases and 10,126 controls as well as 6,113 invasive breast cancer cases from the Breast and Prostate Cancer Cohort Consortium (BPC3). Three SNPs showed at least one nominally significant association in homozygous minor versus homozygous major models. ACVR2A-rs2382112 (ORhom = 3.05, 95%CI = 1.72-5.44, Phom = 1.47 × 10-4 ), MAST2-rs12124649 (ORhom = 1.73, 95% CI =1.18-2.54, Phom = 5.24 × 10-3 ), and INSR-rs10500204 (ORhom = 1.96, 95% CI = 1.44-2.67, Phom =1.68 × 10-5 ) were associated with increased risk of BCIS; however, only the latter association was significant after correcting for multiple testing. Furthermore, INSR-rs10500204 was more strongly associated with the risk of BCIS than invasive disease in case-only analyses using the homozygous minor versus homozygous major model (ORhom = 1.78, 95% CI = 1.30-2.44, Phom = 3.23 × 10-4 ). The SNP INSR-rs10500204 is located in an intron of the INSR gene and is likely to affect binding of the promyelocytic leukemia (PML) protein. The PML gene is known as a tumor suppressor and growth regulator in cancer. However, it is not clear on what pathway the A-allele of rs10500204 could operate to influence the binding of the protein. Hence, functional studies are warranted to investigate this further.
Collapse
Affiliation(s)
- Myrto Barrdahl
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mia M Gaudet
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | | | - Kostas Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece.,Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Carla H van Gils
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Signe Borgquist
- Clinical Trial Unit, Skåne University Hospital, Lund, Sweden.,Division of Oncology and Pathology, Clinical Sciences, Lund, Lund University, Sweden
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway.,Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Genetic Epidemiology Group, Folkhälsan Research Center, Helsinki, Finland
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Graham G Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, VIC, Australia.,Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, VIC, Australia.,Faculty of Medicine, Monash University, Melbourne, VIC, Australia
| | - Roger L Milne
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, VIC, Australia.,Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, VIC, Australia
| | - Loic Le Marchand
- Cancer Research Center of Hawaii, University of Hawaii, Honolulu, HI
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Sara Lindström
- Department of Epidemiology, University of Washington; Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA
| | - David J Hunter
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA
| | - Regina Ziegler
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Pike Bethesda, MD
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Pike Bethesda, MD.,Core Genotyping Facility, Frederick National Laboratory for Cancer Research, Gaithersburg, MD
| | - Xiaohong R Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Pike Bethesda, MD
| | - Julie E Buring
- Divisions of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - I-Min Lee
- Divisions of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
11
|
de Thé H, Pandolfi PP, Chen Z. Acute Promyelocytic Leukemia: A Paradigm for Oncoprotein-Targeted Cure. Cancer Cell 2017; 32:552-560. [PMID: 29136503 DOI: 10.1016/j.ccell.2017.10.002] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/01/2017] [Accepted: 09/29/2017] [Indexed: 12/14/2022]
Abstract
Recent clinical trials have demonstrated that the immense majority of acute promyelocytic leukemia (APL) patients can be definitively cured by the combination of two targeted therapies: retinoic acid (RA) and arsenic. Mouse models have provided unexpected insights into the mechanisms involved. Restoration of PML nuclear bodies upon RA- and/or arsenic-initiated PML/RARA degradation is essential, while RA-triggered transcriptional activation is dispensable for APL eradication. Mutations of the arsenic-binding site of PML/RARA, but also PML, have been detected in therapy-resistant patients, demonstrating the key role of PML in APL cure. PML nuclear bodies are druggable and could be harnessed in other conditions.
Collapse
Affiliation(s)
- Hugues de Thé
- Collège de France, PSL Research University, Chaire d'Oncologie Cellulaire et Moléculaire, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, INSERM UMR 944, CNRS UMR 7212, Hôpital St. Louis, Paris, France.
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zhu Chen
- Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| |
Collapse
|
12
|
Zhu B, Wang JY, Zhou JJ, Zhou F, Cheng W, Liu YT, Wang J, Chen X, Chen DH, Luo L, Hua ZC. PML-RARα stabilized by zinc in human acute promyelocytic leukemia NB4 cells. J Inorg Biochem 2017; 175:92-100. [DOI: 10.1016/j.jinorgbio.2017.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/03/2017] [Accepted: 07/09/2017] [Indexed: 01/09/2023]
|
13
|
Li K, Wang F, Cao WB, Lv XX, Hua F, Cui B, Yu JJ, Zhang XW, Shang S, Liu SS, Yu JM, Han MZ, Huang B, Zhang TT, Li X, Jiang JD, Hu ZW. TRIB3 Promotes APL Progression through Stabilization of the Oncoprotein PML-RARα and Inhibition of p53-Mediated Senescence. Cancer Cell 2017; 31:697-710.e7. [PMID: 28486108 DOI: 10.1016/j.ccell.2017.04.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/10/2016] [Accepted: 04/12/2017] [Indexed: 12/19/2022]
Abstract
Acute promyelocytic leukemia (APL) is driven by the oncoprotein PML-RARα, which antagonizes myeloid differentiation and promotes APL-initiating cell self-renewal. Combined all-trans retinoic acid (ATRA) with arsenic trioxide (As2O3) or chemotherapy dramatically improves the prognosis of APL patients. Here we report that expression of pseudokinase Tribble 3 (TRIB3) associates positively with APL progression and therapeutic resistance. The elevated TRIB3 expression promotes APL by interacting with PML-RARα and suppressing its sumoylation, ubiquitylation, and degradation. This represses PML nuclear body assembly, p53-mediated senescence, and cell differentiation, and supports cellular self-renewal. Genetically inhibiting TRIB3 expression or combination of a peptide disturbing TRIB3/PML-RARα interaction with ATRA/As2O3 eradicates APL by accelerating PML-RARα degradation. Our study provides insight into APL pathogenesis and a potential therapeutic option against APL.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Arsenic Trioxide
- Arsenicals/pharmacology
- Cell Cycle Proteins/deficiency
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Cell Proliferation
- Cellular Senescence
- Disease Progression
- Drug Resistance, Neoplasm
- Female
- Gene Expression Regulation
- Gene Fusion
- HEK293 Cells
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Male
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Oxides/pharmacology
- Peptides/pharmacology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Stability
- Proteolysis
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- Sumoylation
- Time Factors
- Transfection
- Tretinoin/pharmacology
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Ubiquitination
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ke Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Feng Wang
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Wen-Bin Cao
- Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, P.R. China
| | - Xiao-Xi Lv
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Fang Hua
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Bing Cui
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Jiao-Jiao Yu
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Xiao-Wei Zhang
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Shuang Shang
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Shan-Shan Liu
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Jin-Mei Yu
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Ming-Zhe Han
- Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, P.R. China
| | - Bo Huang
- Institute of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Ting-Ting Zhang
- Department of Pharmacy, Marine College, Shandong University, Weihai 264209, P.R. China
| | - Xia Li
- Department of Pharmacy, Marine College, Shandong University, Weihai 264209, P.R. China
| | - Jian-Dong Jiang
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China; Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Zhuo-Wei Hu
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China.
| |
Collapse
|
14
|
Functionally distinct roles for different miR-155 expression levels through contrasting effects on gene expression, in acute myeloid leukaemia. Leukemia 2016; 31:808-820. [PMID: 27740637 DOI: 10.1038/leu.2016.279] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 08/26/2016] [Accepted: 09/06/2016] [Indexed: 12/18/2022]
Abstract
Enforced expression of microRNA-155 (miR-155) in myeloid cells has been shown to have both oncogenic or tumour-suppressor functions in acute myeloid leukaemia (AML). We sought to resolve these contrasting effects of miR-155 overexpression using murine models of AML and human paediatric AML data sets. We show that the highest miR-155 expression levels inhibited proliferation in murine AML models. Over time, enforced miR-155 expression in AML in vitro and in vivo, however, favours selection of intermediate miR-155 expression levels that results in increased tumour burden in mice, without accelerating the onset of disease. Strikingly, we show that intermediate and high miR-155 expression also regulate very different subsets of miR-155 targets and have contrasting downstream effects on the transcriptional environments of AML cells, including genes involved in haematopoiesis and leukaemia. Furthermore, we show that elevated miR-155 expression detected in paediatric AML correlates with intermediate and not high miR-155 expression identified in our experimental models. These findings collectively describe a novel dose-dependent role for miR-155 in the regulation of AML, which may have important therapeutic implications.
Collapse
|
15
|
Dvorkina M, Nieddu V, Chakelam S, Pezzolo A, Cantilena S, Leite AP, Chayka O, Regad T, Pistorio A, Sementa AR, Virasami A, Barton J, Montano X, Lechertier T, Brindle N, Morgenstern D, Lebras M, Burns AJ, Saunders NJ, Hodivala-Dilke K, Bagella L, De The H, Anderson J, Sebire N, Pistoia V, Sala A, Salomoni P. A Promyelocytic Leukemia Protein-Thrombospondin-2 Axis and the Risk of Relapse in Neuroblastoma. Clin Cancer Res 2016; 22:3398-409. [PMID: 27076624 DOI: 10.1158/1078-0432.ccr-15-2081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/19/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Neuroblastoma is a childhood malignancy originating from the sympathetic nervous system with a complex biology, prone to metastasize and relapse. High-risk, metastatic cases are explained in part by amplification or mutation of oncogenes, such as MYCN and ALK, and loss of tumor suppressor genes in chromosome band 1p. However, it is fundamental to identify other pathways responsible for the large portion of neuroblastomas with no obvious molecular alterations. EXPERIMENTAL DESIGN Neuroblastoma cell lines were used for the assessment of tumor growth in vivo and in vitro Protein expression in tissues and cells was assessed using immunofluorescence and IHC. The association of promyelocytic leukemia (PML) expression with neuroblastoma outcome and relapse was calculated using log-rank and Mann-Whitney tests, respectively. Gene expression was assessed using chip microarrays. RESULTS PML is detected in the developing and adult sympathetic nervous system, whereas it is not expressed or is low in metastatic neuroblastoma tumors. Reduced PML expression in patients with low-risk cancers, that is, localized and negative for the MYCN proto-oncogene, is strongly associated with tumor recurrence. PML-I, but not PML-IV, isoform suppresses angiogenesis via upregulation of thrombospondin-2 (TSP2), a key inhibitor of angiogenesis. Finally, PML-I and TSP2 expression inversely correlates with tumor angiogenesis and recurrence in localized neuroblastomas. CONCLUSIONS Our work reveals a novel PML-I-TSP2 axis for the regulation of angiogenesis and cancer relapse, which could be used to identify patients with low-risk, localized tumors that might benefit from chemotherapy. Clin Cancer Res; 22(13); 3398-409. ©2016 AACR.
Collapse
Affiliation(s)
- Maria Dvorkina
- Samantha Dickson Brain Cancer Unit, University College London Cancer Institute, University College London, London, United Kingdom
| | - Valentina Nieddu
- Department of Life Sciences, Institute of Environment and Health, Brunel University London, Uxbridge, United Kingdom. Department of Biomedical Sciences, National Institute of Biostructures and Biosystems, University of Sassari, Sassari, Italy
| | - Shalini Chakelam
- Samantha Dickson Brain Cancer Unit, University College London Cancer Institute, University College London, London, United Kingdom
| | - Annalisa Pezzolo
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Sandra Cantilena
- Department of Life Sciences, Institute of Environment and Health, Brunel University London, Uxbridge, United Kingdom. Laboratorio di Oncologia, Istituto Giannina Gaslini, Genova, Italy
| | - Ana Paula Leite
- Samantha Dickson Brain Cancer Unit, University College London Cancer Institute, University College London, London, United Kingdom
| | - Olesya Chayka
- Samantha Dickson Brain Cancer Unit, University College London Cancer Institute, University College London, London, United Kingdom. UCL Institute of Child Health, London, United Kingdom
| | - Tarik Regad
- Samantha Dickson Brain Cancer Unit, University College London Cancer Institute, University College London, London, United Kingdom. Nottingham Trent University, Nottingham, United Kingdom
| | | | - Angela Rita Sementa
- Laboratorio di Anatomia Patologica, Istituto Giannina Gaslini, Genova, Italy
| | - Alex Virasami
- UCL Institute of Child Health, London, United Kingdom. Epidemiologia e Biostatistica, Istituto Giannina Gaslini, Genova, Italy
| | - Jack Barton
- UCL Institute of Child Health, London, United Kingdom. Epidemiologia e Biostatistica, Istituto Giannina Gaslini, Genova, Italy
| | - Ximena Montano
- UCL Institute of Child Health, London, United Kingdom. Epidemiologia e Biostatistica, Istituto Giannina Gaslini, Genova, Italy
| | | | - Nicola Brindle
- Samantha Dickson Brain Cancer Unit, University College London Cancer Institute, University College London, London, United Kingdom
| | - Daniel Morgenstern
- UCL Institute of Child Health, London, United Kingdom. Epidemiologia e Biostatistica, Istituto Giannina Gaslini, Genova, Italy
| | - Morgane Lebras
- Barts Cancer Institute, Queen Mary University, London, United Kingdom
| | - Alan J Burns
- Laboratorio di Oncologia, Istituto Giannina Gaslini, Genova, Italy. Birth Defects Research Centre. Dept. Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Nigel J Saunders
- Department of Life Sciences, Institute of Environment and Health, Brunel University London, Uxbridge, United Kingdom
| | | | - Luigi Bagella
- Department of Biomedical Sciences, National Institute of Biostructures and Biosystems, University of Sassari, Sassari, Italy. Institut Universitaire d'Hematologie, Sant-Louis Hospital, Paris Diderot University, Paris, France
| | - Hugues De The
- Barts Cancer Institute, Queen Mary University, London, United Kingdom
| | - John Anderson
- UCL Institute of Child Health, London, United Kingdom. Epidemiologia e Biostatistica, Istituto Giannina Gaslini, Genova, Italy
| | - Neil Sebire
- UCL Institute of Child Health, London, United Kingdom. Epidemiologia e Biostatistica, Istituto Giannina Gaslini, Genova, Italy
| | - Vito Pistoia
- Nottingham Trent University, Nottingham, United Kingdom
| | - Arturo Sala
- Department of Life Sciences, Institute of Environment and Health, Brunel University London, Uxbridge, United Kingdom.
| | - Paolo Salomoni
- Samantha Dickson Brain Cancer Unit, University College London Cancer Institute, University College London, London, United Kingdom.
| |
Collapse
|
16
|
Koidl S, Eisenhardt N, Fatouros C, Droescher M, Chaugule VK, Pichler A. The SUMO2/3 specific E3 ligase ZNF451-1 regulates PML stability. Int J Biochem Cell Biol 2016; 79:478-487. [PMID: 27343429 DOI: 10.1016/j.biocel.2016.06.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 01/01/2023]
Abstract
The small ubiquitin related modifier SUMO regulates protein functions to maintain cell homeostasis. SUMO attachment is executed by the hierarchical action of E1, E2 and E3 enzymes of which E3 ligases ensure substrate specificity. We recently identified the ZNF451 family as novel class of SUMO2/3 specific E3 ligases and characterized their function in SUMO chain formation. The founding member, ZNF451isoform1 (ZNF451-1) partially resides in PML bodies, nuclear structures organized by the promyelocytic leukemia gene product PML. As PML and diverse PML components are well known SUMO substrates the question arises whether ZNF451-1 is involved in their sumoylation. Here, we show that ZNF451-1 indeed functions as SUMO2/3 specific E3 ligase for PML and selected PML components in vitro. Mutational analysis indicates that substrate sumoylation employs an identical biochemical mechanism as we described for SUMO chain formation. In vivo, ZNF451-1 RNAi depletion leads to PML stabilization and an increased number of PML bodies. By contrast, PML degradation upon arsenic trioxide treatment is not ZNF451-1 dependent. Our data suggest a regulatory role of ZNF451-1 in fine-tuning physiological PML levels in a RNF4 cooperative manner in the mouse neuroblastoma N2a cell-line.
Collapse
Affiliation(s)
- Stefanie Koidl
- Max Planck Institute of Immunobiology and Epigenetics, Department of Epigenetics, 79108 Freiburg, Stübeweg 51, Germany
| | - Nathalie Eisenhardt
- Max Planck Institute of Immunobiology and Epigenetics, Department of Epigenetics, 79108 Freiburg, Stübeweg 51, Germany
| | - Chronis Fatouros
- Max Planck Institute of Immunobiology and Epigenetics, Department of Epigenetics, 79108 Freiburg, Stübeweg 51, Germany
| | - Mathias Droescher
- Max Planck Institute of Immunobiology and Epigenetics, Department of Epigenetics, 79108 Freiburg, Stübeweg 51, Germany
| | - Viduth K Chaugule
- Max Planck Institute of Immunobiology and Epigenetics, Department of Epigenetics, 79108 Freiburg, Stübeweg 51, Germany
| | - Andrea Pichler
- Max Planck Institute of Immunobiology and Epigenetics, Department of Epigenetics, 79108 Freiburg, Stübeweg 51, Germany.
| |
Collapse
|
17
|
Abstract
The fundamental biological importance of the Tp53 gene family is highlighted by its evolutionary conservation for more than one billion years dating back to the earliest multicellular organisms. The TP53 protein provides essential functions in the cellular response to diverse stresses and safeguards maintenance of genomic integrity, and this is manifest in its critical role in tumor suppression. The importance of Tp53 in tumor prevention is exemplified in human cancer where it is the most frequently detected genetic alteration. This is confirmed in animal models, in which a defective Tp53 gene leads inexorably to cancer development, whereas reinstatement of TP53 function results in regression of established tumors that had been initiated by loss of TP53. Remarkably, despite extensive investigation, the specific mechanisms by which TP53 acts as a tumor suppressor are yet to be fully defined. We review the history and current standing of efforts to understand these mechanisms and how they complement each other in tumor suppression.
Collapse
Affiliation(s)
- Brandon J Aubrey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia Department of Clinical Haematology and Bone Marrow Transplant Service, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| |
Collapse
|
18
|
Guan D, Kao HY. The function, regulation and therapeutic implications of the tumor suppressor protein, PML. Cell Biosci 2015; 5:60. [PMID: 26539288 PMCID: PMC4632682 DOI: 10.1186/s13578-015-0051-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/28/2015] [Indexed: 12/21/2022] Open
Abstract
The tumor suppressor protein, promyelocytic leukemia protein (PML), was originally identified in acute promyelocytic leukemia due to a chromosomal translocation between chromosomes 15 and 17. PML is the core component of subnuclear structures called PML nuclear bodies (PML-NBs), which are disrupted in acute promyelocytic leukemia cells. PML plays important roles in cell cycle regulation, survival and apoptosis, and inactivation or down-regulation of PML is frequently found in cancer cells. More than 120 proteins have been experimentally identified to physically associate with PML, and most of them either transiently or constitutively co-localize with PML-NBs. These interactions are associated with many cellular processes, including cell cycle arrest, apoptosis, senescence, transcriptional regulation, DNA repair and intermediary metabolism. Importantly, PML inactivation in cancer cells can occur at the transcriptional-, translational- or post-translational- levels. However, only a few somatic mutations have been found in cancer cells. A better understanding of its regulation and its role in tumor suppression will provide potential therapeutic opportunities. In this review, we discuss the role of PML in multiple tumor suppression pathways and summarize the players and stimuli that control PML protein expression or subcellular distribution.
Collapse
Affiliation(s)
- Dongyin Guan
- Department of Biochemistry, School of Medicine, Case Western Reserve University, and Comprehensive Cancer Center of Case Western Reserve University, Cleveland, 10900 Euclid Avenue, Cleveland, OH 44106 USA
| | - Hung-Ying Kao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, and Comprehensive Cancer Center of Case Western Reserve University, Cleveland, 10900 Euclid Avenue, Cleveland, OH 44106 USA
| |
Collapse
|
19
|
Yu M, Wang J, Ma D, Chen S, Lin X, Fang Q, Zhe N. HO-1, RET and PML as possible markers for risk stratification of acute myelocytic leukemia and prognostic evaluation. Oncol Lett 2015; 10:3137-3144. [PMID: 26722301 DOI: 10.3892/ol.2015.3644] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 07/28/2015] [Indexed: 11/06/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible isoform of HO that is activated in response to oxidative stress and has anti-apoptotic and pro-proliferative effects on leukemia cells. RET, a tyrosine kinase receptor; its expression levels are associated with the differentiation degree of acute myelocytic leukemia (AML) cells. The promyelocytic leukemia (PML) gene inhibits cell proliferation and tumor growth, participates in the differentiation of hematopoietic progenitor cells and induces cell apoptosis. However, the association between the expression levels of HO-1, RET and PML genes and the risk stratification of AML and prognosis have not previously been reported. In the present study, HO-1 was expressed in the human AML Kasumi-1, HL-60 and THP-1 cell lines, and HO-1 expression was regulated by Hemin (20 µmol/l) and ZnPPIX (10 µmol/l). Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis demonstrated that expression of RET and PML were positively and negatively correlated with HO-1 expression, respectively. Bone marrow samples (18 favorable, 55 intermediate, 15 adverse and 2 unknown karyotype AML cases and 20 healthy donors) were collected from 90 randomly selected AML patients upon their first visit. The mRNA and protein expression of HO-1, RET and PML in samples was detected by RT-qPCR and western blot analysis. At the mRNA level, the adverse group expressed significantly higher levels of HO-1 and RET compared with the levels in the favorable and normal groups. The PML mRNA expression levels in adverse patient samples was lower compared with those of the intermediate group and favorable group. Western blot analysis demonstrated that the expression levels of HO-1, RET and PML proteins in all risk groups exhibited the same pattern of expression as was observed for the mRNA levels. The overall survival and relapse-free survival rates were shortest in AML patients with high HO-1 expression (Kaplan-Meier; log-rank, P<0.01). The results of the present study therefore indicate that HO-1, RET and PML may be critical in the risk-stratification and prognosis of AML. However, additional samples and clinical data should be collected and analyzed in order to provide stronger evidence for this hypothesis.
Collapse
Affiliation(s)
- Meisheng Yu
- Clinical Medical College, Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Jishi Wang
- Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China ; Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Dan Ma
- Clinical Medical College, Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China ; Department of Pharmacy, Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Shuya Chen
- Clinical Medical College, Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Xiaojing Lin
- Clinical Medical College, Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Qin Fang
- Department of Pharmacy, Affiliated Baiyun Hospital of Guiyang Medical University, Guiyang, Guizhou 550058, P.R. China
| | - Nana Zhe
- Clinical Medical College, Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
20
|
Wang Y, Jin W, Jia X, Luo R, Tan Y, Zhu X, Yang X, Wang X, Wang K. Transcriptional repression of CDKN2D by PML/RARα contributes to the altered proliferation and differentiation block of acute promyelocytic leukemia cells. Cell Death Dis 2014; 5:e1431. [PMID: 25275592 PMCID: PMC4649503 DOI: 10.1038/cddis.2014.388] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 07/09/2014] [Accepted: 07/14/2014] [Indexed: 01/14/2023]
Abstract
Cell proliferation and differentiation are highly coordinated processes. These two processes are disrupted during leukemogenesis, resulting in differentiation block and uncontrolled proliferation in leukemia. To understand the mechanisms disrupting the coordination between the two processes in acute promyelocytic leukemia (APL), we investigated the regulatory mechanism of the negative cell cycle regulator CDKN2D by the promyelocytic leukemia/retinoic acid receptor α (PML/RARα) fusion protein and the role of CDKN2D in cell differentiation and proliferation. We found that CDKN2D expression in APL cells was significantly lower than that in normal promyelocytes. By chromatin immunoprecipitation and luciferase reporter assays, we showed that PML/RARα directly bound to and inhibited the transactivation of the CDKN2D promoter. Further evidence by the truncated and mutated CDKN2D promoters revealed that the everted repeat 8 (ER8) motif on the promoter was the binding site of PML/RARα. Forced expression of CDKN2D induced G0/G1 phase arrest and partial granulocytic differentiation in APL-derived NB4 cells, suggesting the function of CDKN2D in regulating both cell proliferation and granulocytic differentiation. Furthermore, all-trans retinoic acid (ATRA) significantly induced CDKN2D expression in APL cells and knockdown of CDKN2D expression during ATRA treatment partially blocked the ATRA-induced differentiation and cell cycle arrest. Collectively, our data indicate that CDKN2D repression by PML/RARα disrupts both cell proliferation and differentiation in the pathogenesis of APL, and induced expression of CDKN2D by ATRA alleviates the disruption of both processes to ensure treatment efficiency. This study provides a mechanism for coupling proliferation and differentiation in leukemic cells through the action of CDKN2D.
Collapse
Affiliation(s)
- Y Wang
- State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - W Jin
- 1] State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China [2] Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, China [3] Sino-French Research Center for Life Sciences and Genomics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - X Jia
- State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - R Luo
- School of Life Sciences/Center for Computational Systems Biology, Fudan University, Shanghai, 200433, China
| | - Y Tan
- State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - X Zhu
- State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - X Yang
- State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - X Wang
- State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - K Wang
- 1] State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China [2] Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, China [3] Sino-French Research Center for Life Sciences and Genomics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
21
|
Abstract
p53 is a crucial tumour suppressor that responds to diverse stress signals by orchestrating specific cellular responses, including transient cell cycle arrest, cellular senescence and apoptosis, which are all processes associated with tumour suppression. However, recent studies have challenged the relative importance of these canonical cellular responses for p53-mediated tumour suppression and have highlighted roles for p53 in modulating other cellular processes, including metabolism, stem cell maintenance, invasion and metastasis, as well as communication within the tumour microenvironment. In this Opinion article, we discuss the roles of classical p53 functions, as well as emerging p53-regulated processes, in tumour suppression.
Collapse
Affiliation(s)
- Kathryn T Bieging
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, CCSR-South, Room 1255, 269 Campus Drive, Stanford, California 94305, USA
| | - Stephano Spano Mello
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, CCSR-South, Room 1255, 269 Campus Drive, Stanford, California 94305, USA
| | - Laura D Attardi
- 1] Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, CCSR-South, Room 1255, 269 Campus Drive, Stanford, California 94305, USA. [2] Department of Genetics, Stanford University School of Medicine, CCSR-South, Room 1255, 269 Campus Drive, Stanford, California 94305, USA
| |
Collapse
|
22
|
Münch S, Weidtkamp-Peters S, Klement K, Grigaravicius P, Monajembashi S, Salomoni P, Pandolfi PP, Weißhart K, Hemmerich P. The tumor suppressor PML specifically accumulates at RPA/Rad51-containing DNA damage repair foci but is nonessential for DNA damage-induced fibroblast senescence. Mol Cell Biol 2014; 34:1733-46. [PMID: 24615016 PMCID: PMC4019039 DOI: 10.1128/mcb.01345-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/19/2013] [Accepted: 02/14/2014] [Indexed: 12/24/2022] Open
Abstract
The PML tumor suppressor has been functionally implicated in DNA damage response and cellular senescence. Direct evidence for such a role based on PML knockdown or knockout approaches is still lacking. We have therefore analyzed the irradiation-induced DNA damage response and cellular senescence in human and mouse fibroblasts lacking PML. Our data show that PML nuclear bodies (NBs) nonrandomly associate with persistent DNA damage foci in unperturbed human skin and in high-dose-irradiated cell culture systems. PML bodies do not associate with transient γH2AX foci after low-dose gamma irradiation. Superresolution microscopy reveals that all PML bodies within a nucleus are engaged at Rad51- and RPA-containing repair foci during ongoing DNA repair. The lack of PML (i) does not majorly affect the DNA damage response, (ii) does not alter the efficiency of senescence induction after DNA damage, and (iii) does not affect the proliferative potential of primary mouse embryonic fibroblasts during serial passaging. Thus, while PML NBs specifically accumulate at Rad51/RPA-containing lesions and senescence-derived persistent DNA damage foci, they are not essential for DNA damage-induced and replicative senescence of human and murine fibroblasts.
Collapse
Affiliation(s)
- Sandra Münch
- Leibniz Institute for Age Research, Jena, Germany
| | | | | | | | | | - Paolo Salomoni
- University College London, UCL Cancer Institute, London, United Kingdom
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Klaus Weißhart
- Carl Zeiss Microscopy GmbH, BioSciences Division, Jena, Germany
| | | |
Collapse
|
23
|
Sinclair A, Latif AL, Holyoake TL. Targeting survival pathways in chronic myeloid leukaemia stem cells. Br J Pharmacol 2014; 169:1693-707. [PMID: 23517124 DOI: 10.1111/bph.12183] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 02/11/2013] [Accepted: 02/19/2013] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Chronic myeloid leukaemia (CML) is a clonal myeloproliferative disorder characterized by the presence of a fusion oncogene BCR-ABL, which encodes a protein with constitutive TK activity. The implementation of tyrosine kinase inhibitors (TKIs) marked a major advance in CML therapy; however, there are problems with current treatment. For example, relapse occurs when these drugs are discontinued in the majority of patients who have achieved a complete molecular response on TKI and these agents are less effective in patients with mutations in the BCR-ABL kinase domain. Importantly, TKI can effectively target proliferating mature cells, but do not eradicate quiescent leukaemic stem cells (LSCs), therefore allowing disease persistence despite treatment. It is essential that alternative strategies are used to target the LSC population. BCR-ABL activation is responsible for the modulation of different signalling pathways, which allows the LSC fraction to evade cell death. Several pathways have been shown to be modulated by BCR-ABL, including PI3K/AKT/mTOR, JAK-STAT and autophagy signalling pathways. Targeting components of these survival pathways, alone or in combination with TKI, therefore represents an attractive potential therapeutic approach for targeting the LSC. However, many pathways are also active in normal stem cells. Therefore, potential targets must be validated to effectively eradicate CML stem cells while sparing normal counterparts. This review summarizes the main pathways modulated in CML stem cells, the recent developments and the use of novel drugs to target components in these pathways which may be used to target the LSC population. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
Affiliation(s)
- A Sinclair
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | | | | |
Collapse
|
24
|
Abstract
The role of the promyelocytic leukemia (PML) protein has been widely tested in many different contexts, as attested by the hundreds of papers present in the literature. In most of these studies, PML is regarded as a tumor suppressor, a notion on the whole accepted by the scientific community. In this review, we examine how the concept of tumor-suppressor gene has evolved until now and then systematically assess whether this assumption for PML is supported by unambiguous experimental evidence.
Collapse
Affiliation(s)
- Massimiliano Mazza
- Department of Experimental Oncology, European Institute of Oncology , Milan , Italy
| | | |
Collapse
|
25
|
Haupt S, Mitchell C, Corneille V, Shortt J, Fox S, Pandolfi PP, Castillo-Martin M, Bonal DM, Cordon-Cardo C, Lozano G, Haupt Y. Loss of PML cooperates with mutant p53 to drive more aggressive cancers in a gender-dependent manner. Cell Cycle 2013; 12:1722-31. [PMID: 23656786 DOI: 10.4161/cc.24805] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED p53 mutations and downregulation of promyelocytic leukemia (PML) are common genetic alterations in human cancers. In healthy cells these two key tumor suppressors exist in a positive regulatory loop, promoting cell death and cellular senescence. However, the influence of their interplay on tumorigenesis has not been explored directly in vivo. The contribution of PML to mutant p53 driven cancer was evaluated in a mouse model harboring a p53 mutation (p53 (wild-type/R172H) ) that recapitulates a frequent p53 mutation (p53 (R175H) ) in human sporadic and Li-Fraumeni cancers. These mice with PML displayed perturbation of the hematopoietic compartment, manifested either as lymphoma or extramedullary hematopoiesis (EMH). EMH was associated with peripheral blood leucocytosis and macrocytic anemia, suggestive of myeloproliferative- myelodysplastic overlap. In contrast, a complete loss of PML from these mice resulted in a marked alteration in tumor profile. While the incidence of lymphomas was unaltered, EMH was not detected and the majority of mice succumbed to sarcomas. Further, males lacking PML exhibited a high incidence of soft tissue sarcomas and reduced survival, while females largely developed osteosarcomas, without impact on survival. Together, these findings demonstrate that PML is an important tumor suppressor dictating disease development in a pertinent mouse model of human cancer. KEY POINTS (1) A mutant p53 allele disrupts hematopoiesis in mice, by promoting lymphomas and myeloproliferative / myelodysplastic overlap. (2) Coincidental p53 allele mutation and PML loss shifts the tumor profile toward sarcoma formation, which is paralleled in human leiomyosarcomas (indicated by immunohistochemistry; IHC).
Collapse
Affiliation(s)
- Sue Haupt
- Research Division, Peter MacCallum Cancer Centre, East Melbourne, VIC Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Rabellino A, Scaglioni PP. PML Degradation: Multiple Ways to Eliminate PML. Front Oncol 2013; 3:60. [PMID: 23526763 PMCID: PMC3605509 DOI: 10.3389/fonc.2013.00060] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 03/06/2013] [Indexed: 11/26/2022] Open
Abstract
The promyelocytic leukemia tumor suppressor gene (PML) critically regulates several cellular functions that oppose tumorigenesis such as oncogene-induced senescence, apoptosis, the response to DNA damage and to viral infections. PML deficiency occurs commonly in a broad spectrum of human cancers through mechanisms that involve its aberrant ubiquitination and degradation. Furthermore, several viruses encode viral proteins that promote viral replication through degradation of PML. These observations suggest that restoration of PML should lead to potent antitumor effects or antiviral responses. In this review we will summarize the mechanisms involved in PML degradation with the intent to highlight novel therapeutic strategies to trigger PML restoration.
Collapse
Affiliation(s)
- Andrea Rabellino
- Division of Hematology and Oncology, Department of Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center Dallas, TX, USA
| | | |
Collapse
|
27
|
Activation of protein kinase CK2 attenuates FOXO3a functioning in a PML-dependent manner: implications in human prostate cancer. Cell Death Dis 2013; 4:e543. [PMID: 23492774 PMCID: PMC3613841 DOI: 10.1038/cddis.2013.63] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Protein kinase CK2 (also known as Caseine Kinase II) is an ubiquitous Ser/Thr protein kinase present in both the nucleus and cytoplasm of cells, targeting several key enzymes, growth factor receptors, transcription factors and cytoskeletal proteins. It is not only a key player in regulating cellular growth and proliferation, but also behaves as a potent suppressor of apoptosis. CK2 has been frequently found to be deregulated (mostly hyperactivated) in all cancers, prostate cancer being prominent of them. In the recent past, tumor suppressor PML (promyelocytic leukemia) has been shown to be a target of phosphorylation by CK2. This phosphorylation promotes the ubiquitin-mediated proteasomal degradation of PML thereby effectively curbing its role as a tumor suppressor. Among many others, PML has also been established to mediate its tumor suppressive role by mitigating the inactivation of active AKT (pAKT) inside the nucleus by assembling a dephosphorylating platform for nuclear pAKT. One of the immediate consequences, of this inactivation is the stabilization of FOXO3a, another well-established tumor suppressor, inside the nucleus and its downstream activities. Here, we propose a novel signaling axis apexed by deregulated CK2, dismantling the association of PML and PHLPP2 (we also report PHLPP2 to be a novel interacting partner of PML inside the nucleus), ultimately leading to the inactivation and nuclear exclusion of FOXO3a, thereby downregulating p21/p27/Bim in which degradation of PML and the concomitant stabilization of pAKT plays a cardinal part.
Collapse
|
28
|
Butler K, Martinez LA, Tejada-Simon MV. Impaired cognitive function and reduced anxiety-related behavior in a promyelocytic leukemia (PML) tumor suppressor protein-deficient mouse. GENES BRAIN AND BEHAVIOR 2013; 12:189-202. [PMID: 23279884 DOI: 10.1111/gbb.12014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 08/03/2012] [Accepted: 12/19/2012] [Indexed: 01/23/2023]
Abstract
The promyelocytic leukemia (PML) protein is a tumor suppressor factor mostly known by its involvement in acute promyelocytic leukemia (APL). Interestingly, recent studies have provided evidence that, in the central nervous system, PML is involved in neurogenesis. However, prospective studies of PML in brain are lacking. To further understand the role of PML in the mammalian brain, we studied plasticity and behavioral changes in PML knockout mice. If PML is involved in neurogenesis, and neurogenesis is an important process for proper brain development as well as learning and memory functions, we hypothesized that PML might have a role in plasticity and cognition. Behavioral studies demonstrated that PML knockout mice present abnormalities in conditioned learning and spatial memory, as determined by fear conditioning and Morris water maze tasks. Experiments to determine normal exploratory behavior interestingly revealed that PML knockout mice present reduced anxiety-related responses as compared to control animals. This was confirmed when PML knockout mice spent more time in the open arms of an elevated plus-maze, which is an indication of decreased anxiety. Additionally, impairments in hippocampus-dependent learning were mirrored by altered long-term plasticity at Schaffer collateral-CA1 synapses. We now provide the first evidence for an important role of PML in the brain, indicating that PML might have a role in synaptic plasticity and associated behavioral processes.
Collapse
Affiliation(s)
- K Butler
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | | | | |
Collapse
|
29
|
Akl H, Bultynck G. Altered Ca(2+) signaling in cancer cells: proto-oncogenes and tumor suppressors targeting IP3 receptors. Biochim Biophys Acta Rev Cancer 2012; 1835:180-93. [PMID: 23232185 DOI: 10.1016/j.bbcan.2012.12.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/30/2012] [Accepted: 12/01/2012] [Indexed: 01/15/2023]
Abstract
Proto-oncogenes and tumor suppressors critically control cell-fate decisions like cell survival, adaptation and death. These processes are regulated by Ca(2+) signals arising from the endoplasmic reticulum, which at distinct sites is in close proximity to the mitochondria. These organelles are linked by different mechanisms, including Ca(2+)-transport mechanisms involving the inositol 1,4,5-trisphosphate receptor (IP3R) and the voltage-dependent anion channel (VDAC). The amount of Ca(2+) transfer from the endoplasmic reticulum to mitochondria determines the susceptibility of cells to apoptotic stimuli. Suppressing the transfer of Ca(2+) from the endoplasmic reticulum to the mitochondria increases the apoptotic resistance of cells and may decrease the cellular responsiveness to apoptotic signaling in response to cellular damage or alterations. This can result in the survival, growth and proliferation of cells with oncogenic features. Clearly, proper maintenance of endoplasmic reticulum Ca(2+) homeostasis and dynamics including its links with the mitochondrial network is essential to detect and eliminate altered cells with oncogenic features through the apoptotic pathway. Proto-oncogenes and tumor suppressors exploit the central role of Ca(2+) signaling by targeting the IP3R. There are an increasing number of reports showing that activation of proto-oncogenes or inactivation of tumor suppressors directly affects IP3R function and endoplasmic reticulum Ca(2+) homeostasis, thereby decreasing mitochondrial Ca(2+) uptake and mitochondrial outer membrane permeabilization. In this review, we provide an overview of the current knowledge on the proto-oncogenes and tumor suppressors identified as IP3R-regulatory proteins and how they affect endoplasmic reticulum Ca(2+) homeostasis and dynamics.
Collapse
Affiliation(s)
- Haidar Akl
- Department Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
30
|
Bassi SC, Rego EM. Molecular basis for the diagnosis and treatment of acute promyelocytic leukemia. Rev Bras Hematol Hemoter 2012; 34:134-9. [PMID: 23049403 PMCID: PMC3459394 DOI: 10.5581/1516-8484.20120033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 01/31/2012] [Indexed: 11/27/2022] Open
Abstract
Acute promyelocytic leukemia is characterized by gene rearrangements that always involve the retinoic acid receptor alpha on chromosome 15. In the majority of patients t(15;17) is detected, which generates the promyelocytic leukemia gene/retinoic acid receptor alpha rearrangement. This rearrangement interacts with several proteins, including the native promyelocytic leukemia gene, thus causing its delocalization from the nuclear bodies, impairing its function. The immunofluorescence staining technique using the anti-PML antibody may be used to provide a rapid diagnosis and to immediately start therapy using all-trans retinoic acid. The experience of the International Consortium on Acute Promyelocytic Leukemia has demonstrated that early mortality was significantly reduced by adopting the immunofluorescence technique. All-trans retinoic acid combined with chemotherapy is the standard therapy; this promotes complete remission rates greater than 90% and cure rates of nearly 80%. However, early mortality is still an important limitation and hematologists must be aware of the importance of treating newly diagnosed acute promyelocytic leukemia as a medical emergency.
Collapse
Key Words
- Leukemia, myeloid, acute/diagnosis
- Leukemia, myeloid, acute/drug therapy
- Leukemia, myeloid, acute/epidemiology
- Leukemia, promyelocytic, acute/diagnosis
- Leukemia, promyelocytic, acute/drug therapy
- Leukemia, promyelocytic, acute/epidemilogy
Collapse
Affiliation(s)
- Sarah Cristina Bassi
- Hematology and Oncology Division, Medical School of Ribeirão Preto, Universidade de São Paulo - USP, Ribeirão Preto, SP, Brazil
| | | |
Collapse
|
31
|
Expression and function of PML-RARA in the hematopoietic progenitor cells of Ctsg-PML-RARA mice. PLoS One 2012; 7:e46529. [PMID: 23056333 PMCID: PMC3466302 DOI: 10.1371/journal.pone.0046529] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 09/05/2012] [Indexed: 12/26/2022] Open
Abstract
Because PML-RARA-induced acute promyelocytic leukemia (APL) is a morphologically differentiated leukemia, many groups have speculated about whether its leukemic cell of origin is a committed myeloid precursor (e.g. a promyelocyte) versus an hematopoietic stem/progenitor cell (HSPC). We originally targeted PML-RARA expression with CTSG regulatory elements, based on the early observation that this gene was maximally expressed in cells with promyelocyte morphology. Here, we show that both Ctsg, and PML-RARA targeted to the Ctsg locus (in Ctsg-PML-RARA mice), are expressed in the purified KLS cells of these mice (KLS = Kit+Lin−Sca+, which are highly enriched for HSPCs), and this expression results in biological effects in multi-lineage competitive repopulation assays. Further, we demonstrate the transcriptional consequences of PML-RARA expression in Ctsg-PML-RARA mice in early myeloid development in other myeloid progenitor compartments [common myeloid progenitors (CMPs) and granulocyte/monocyte progenitors (GMPs)], which have a distinct gene expression signature compared to wild-type (WT) mice. Although PML-RARA is indeed expressed at high levels in the promyelocytes of Ctsg-PML-RARA mice and alters the transcriptional signature of these cells, it does not induce their self-renewal. In sum, these results demonstrate that in the Ctsg-PML-RARA mouse model of APL, PML-RARA is expressed in and affects the function of multipotent progenitor cells. Finally, since PML/Pml is normally expressed in the HSPCs of both humans and mice, and since some human APL samples contain TCR rearrangements and express T lineage genes, we suggest that the very early hematopoietic expression of PML-RARA in this mouse model may closely mimic the physiologic expression pattern of PML-RARA in human APL patients.
Collapse
|
32
|
Welch JS, Ley TJ, Link DC, Miller CA, Larson DE, Koboldt DC, Wartman LD, Lamprecht TL, Liu F, Xia J, Kandoth C, Fulton RS, McLellan MD, Dooling DJ, Wallis JW, Chen K, Harris CC, Schmidt HK, Kalicki-Veizer JM, Lu C, Zhang Q, Lin L, O'Laughlin MD, McMichael JF, Delehaunty KD, Fulton LA, Magrini VJ, McGrath SD, Demeter RT, Vickery TL, Hundal J, Cook LL, Swift GW, Reed JP, Alldredge PA, Wylie TN, Walker JR, Watson MA, Heath SE, Shannon WD, Varghese N, Nagarajan R, Payton JE, Baty JD, Kulkarni S, Klco JM, Tomasson MH, Westervelt P, Walter MJ, Graubert TA, DiPersio JF, Ding L, Mardis ER, Wilson RK. The origin and evolution of mutations in acute myeloid leukemia. Cell 2012; 150:264-78. [PMID: 22817890 DOI: 10.1016/j.cell.2012.06.023] [Citation(s) in RCA: 1239] [Impact Index Per Article: 95.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 04/27/2012] [Accepted: 06/24/2012] [Indexed: 10/28/2022]
Abstract
Most mutations in cancer genomes are thought to be acquired after the initiating event, which may cause genomic instability and drive clonal evolution. However, for acute myeloid leukemia (AML), normal karyotypes are common, and genomic instability is unusual. To better understand clonal evolution in AML, we sequenced the genomes of M3-AML samples with a known initiating event (PML-RARA) versus the genomes of normal karyotype M1-AML samples and the exomes of hematopoietic stem/progenitor cells (HSPCs) from healthy people. Collectively, the data suggest that most of the mutations found in AML genomes are actually random events that occurred in HSPCs before they acquired the initiating mutation; the mutational history of that cell is "captured" as the clone expands. In many cases, only one or two additional, cooperating mutations are needed to generate the malignant founding clone. Cells from the founding clone can acquire additional cooperating mutations, yielding subclones that can contribute to disease progression and/or relapse.
Collapse
Affiliation(s)
- John S Welch
- Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
de Thé H, Le Bras M, Lallemand-Breitenbach V. The cell biology of disease: Acute promyelocytic leukemia, arsenic, and PML bodies. J Cell Biol 2012; 198:11-21. [PMID: 22778276 PMCID: PMC3392943 DOI: 10.1083/jcb.201112044] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 06/15/2012] [Indexed: 12/12/2022] Open
Abstract
Acute promyelocytic leukemia (APL) is driven by a chromosomal translocation whose product, the PML/retinoic acid (RA) receptor α (RARA) fusion protein, affects both nuclear receptor signaling and PML body assembly. Dissection of APL pathogenesis has led to the rediscovery of PML bodies and revealed their role in cell senescence, disease pathogenesis, and responsiveness to treatment. APL is remarkable because of the fortuitous identification of two clinically effective therapies, RA and arsenic, both of which degrade PML/RARA oncoprotein and, together, cure APL. Analysis of arsenic-induced PML or PML/RARA degradation has implicated oxidative stress in the biogenesis of nuclear bodies and SUMO in their degradation.
Collapse
Affiliation(s)
- Hugues de Thé
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 944, Equipe labellisée par la Ligue Nationale contre le Cancer, 2 University Paris-Diderot, Sorbonne Paris Cité, Paris, France.
| | | | | |
Collapse
|
34
|
Cheng X, Liu Y, Chu H, Kao HY. Promyelocytic leukemia protein (PML) regulates endothelial cell network formation and migration in response to tumor necrosis factor α (TNFα) and interferon α (IFNα). J Biol Chem 2012; 287:23356-67. [PMID: 22589541 DOI: 10.1074/jbc.m112.340505] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Promyelocytic leukemia protein (PML) is a tumor suppressor that is highly expressed in vascular endothelium and inflamed tissues, yet its role in inflammation-associated cytokine-regulated angiogenesis and underlying mechanism remains largely unclear. We show that tumor necrosis factor α (TNFα) and interferon α (IFNα) stimulate PML expression while suppressing EC network formation and migration, two key events during angiogenesis. By a knockdown approach, we demonstrate that PML is indispensable for TNFα- and IFNα-mediated inhibition of EC network formation. We further demonstrate that signal transducer and activator of transcription 1 (STAT1) binds PML promoter and that is an important regulator of PML expression. Knockdown of STAT1 reduces endogenous PML and blocks TNFα- and IFNα-induced PML accumulation and relieves TNFα- and IFNα-mediated inhibition of EC network formation. Our data also indicate that PML regulates EC migration, in part, by modulating expression of downstream genes, such as negatively regulating integrin β1 (ITGB1). In addition, knockdown of STAT1 or PML alleviates TNFα- and IFNα-mediated inhibition of ITGB1 expression. Antibody blockade demonstrates that ITGB1 is functionally important for PML- and STAT1-regulated EC migration. Taken together, our data provide novel mechanistic insights that PML functions as a negative regulator in EC network formation and migration.
Collapse
Affiliation(s)
- Xiwen Cheng
- Department of Biochemistry, School of Medicine, Case Western Reserve University and the Comprehensive Cancer Center of CWRU and University Hospital of Cleveland, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
35
|
Giorgi C, Baldassari F, Bononi A, Bonora M, De Marchi E, Marchi S, Missiroli S, Patergnani S, Rimessi A, Suski JM, Wieckowski MR, Pinton P. Mitochondrial Ca(2+) and apoptosis. Cell Calcium 2012; 52:36-43. [PMID: 22480931 PMCID: PMC3396846 DOI: 10.1016/j.ceca.2012.02.008] [Citation(s) in RCA: 359] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 02/23/2012] [Accepted: 02/24/2012] [Indexed: 01/13/2023]
Abstract
Mitochondria are key decoding stations of the apoptotic process. In support of this view, a large body of experimental evidence has unambiguously revealed that, in addition to the well-established function of producing most of the cellular ATP, mitochondria play a fundamental role in triggering apoptotic cell death. Various apoptotic stimuli cause the release of specific mitochondrial pro-apoptotic factors into the cytosol. The molecular mechanism of this release is still controversial, but there is no doubt that mitochondrial calcium (Ca(2+)) overload is one of the pro-apoptotic ways to induce the swelling of mitochondria, with perturbation or rupture of the outer membrane, and in turn the release of mitochondrial apoptotic factors into the cytosol. Here, we review as different proteins that participate in mitochondrial Ca(2+) homeostasis and in turn modulate the effectiveness of Ca(2+)-dependent apoptotic stimuli. Strikingly, the final outcome at the cellular level is similar, albeit through completely different molecular mechanisms: a reduced mitochondrial Ca(2+) overload upon pro-apoptotic stimuli that dramatically blunts the apoptotic response.
Collapse
Affiliation(s)
- Carlotta Giorgi
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Shi L, Oberdoerffer P. Chromatin dynamics in DNA double-strand break repair. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1819:811-9. [PMID: 22285574 DOI: 10.1016/j.bbagrm.2012.01.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 12/28/2011] [Accepted: 01/05/2012] [Indexed: 12/29/2022]
Abstract
DNA double-strand breaks (DSBs) occur in the context of a highly organized chromatin environment and are, thus, a significant threat to the epigenomic integrity of eukaryotic cells. Changes in break-proximal chromatin structure are thought to be a prerequisite for efficient DNA repair and may help protect the structural integrity of the nucleus. Unlike most bona fide DNA repair factors, chromatin influences the repair process at several levels: the existing chromatin context at the site of damage directly affects the access and kinetics of the repair machinery; DSB induced chromatin modifications influence the choice of repair factors, thereby modulating repair outcome; lastly, DNA damage can have a significant impact on chromatin beyond the site of damage. We will discuss recent findings that highlight both the complexity and importance of dynamic and tightly orchestrated chromatin reorganization to ensure efficient DSB repair and nuclear integrity. This article is part of a Special Issue entitled: Chromatin in time and space.
Collapse
Affiliation(s)
- Lei Shi
- Mouse Cancer Genetics Program, NCI- Frederick, NIH, Frederick, MD 21702, USA
| | | |
Collapse
|
37
|
Abstract
The promyelocytic leukaemia gene PML was originally identified at the t(15;17) translocation of acute promyelocytic leukaemia, which generates the oncogene PML-retinoic acid receptor α. PML epitomises a subnuclear structure called PML nuclear body. Current models propose that PML through its scaffold properties is able to control cell growth and survival at many different levels. Here we discuss the current literature and propose new avenues for investigation.
Collapse
|
38
|
Curing APL through PML/RARA degradation by As2O3. Trends Mol Med 2012; 18:36-42. [DOI: 10.1016/j.molmed.2011.10.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/03/2011] [Accepted: 10/05/2011] [Indexed: 11/19/2022]
|
39
|
TRIM involvement in transcriptional regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 770:59-76. [PMID: 23631000 DOI: 10.1007/978-1-4614-5398-7_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Members of the tripartite motif (TRIM) protein family are found in all multicellular eukaryotes and function in a wide range of cellular processes such as cell cycle regulation, differentiation, development, oncogenesis and viral response. Over the past few years, several TRIM proteins have been reported to control gene expression through regulation of the transcriptional activity of numerous sequence-specific transcription factors. These proteins include the transcriptional intermediary factor 1 (TIF1) regulators, the promyelocytic leukemia tumor suppressor PML and the RET finger protein (RFP). In this chapter, we will consider the molecular interactions made by these TRIM proteins and will attempt to clarify some of the molecular mechanisms underlying their regulatory effect on transcription.
Collapse
|
40
|
Bieging KT, Attardi LD. Deconstructing p53 transcriptional networks in tumor suppression. Trends Cell Biol 2011; 22:97-106. [PMID: 22154076 DOI: 10.1016/j.tcb.2011.10.006] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 10/26/2011] [Accepted: 10/27/2011] [Indexed: 12/16/2022]
Abstract
p53 is a pivotal tumor suppressor that induces apoptosis, cell-cycle arrest and senescence in response to stress signals. Although p53 transcriptional activation is important for these responses, the mechanisms underlying tumor suppression have been elusive. To date, no single or compound mouse knockout of specific p53 target genes has recapitulated the dramatic tumor predisposition that characterizes p53-null mice. Recently, however, analysis of knock-in mice expressing p53 transactivation domain mutants has revealed a group of primarily novel direct p53 target genes that may mediate tumor suppression in vivo. We present here an overview of well-known p53 target genes and the tumor phenotypes of the cognate knockout mice, and address the recent identification of new p53 transcriptional targets and how they enhance our understanding of p53 transcriptional networks central for tumor suppression.
Collapse
Affiliation(s)
- Kathryn T Bieging
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|
41
|
Giorgi C, Wieckowski MR, Pandolfi PP, Pinton P. Mitochondria associated membranes (MAMs) as critical hubs for apoptosis. Commun Integr Biol 2011; 4:334-5. [PMID: 21980573 DOI: 10.4161/cib.4.3.15021] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Accepted: 01/30/2011] [Indexed: 11/19/2022] Open
Abstract
Apoptosis is a process of major biomedical interest, since its deregulation is involved in the pathogenesis of a broad variety of disorders (neoplasia, autoimmune disorders, viral and neurodegenerative diseases, to name a few). It is now firmly established that variations in cellular calcium (Ca(2+)) concentration are pivotal in the control of a variety of cellular functions. Strong evidence has been accumulated supporting a central role of Ca(2+) in the regulation of cell death. In particular, in the context of the biochemical mechanisms of apoptosis, increasing evidence support a role for endoplasmic reticulum (ER)-mitochondria Ca(2+) cross talk as a crucial regulator of several pathways of apoptosis. Recent data highlight as also the promyelocytic leukemia protein (PML), by modulating the ER machinery at the contact sites between ER and mitochondria (the mitochondria associated membranes, MAMs), regulates cell survival through the ER-cytosol/mitochondria Ca(2+) signaling.
Collapse
Affiliation(s)
- Carlotta Giorgi
- Department of Experimental and Diagnostic Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI) and LTTA Center; University of Ferrara; Ferrara, Italy
| | | | | | | |
Collapse
|
42
|
Welch JS, Yuan W, Ley TJ. PML-RARA can increase hematopoietic self-renewal without causing a myeloproliferative disease in mice. J Clin Invest 2011; 121:1636-45. [PMID: 21364283 DOI: 10.1172/jci42953] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 01/05/2011] [Indexed: 01/20/2023] Open
Abstract
Acute promyelocytic leukemia (APL) is characterized by the t(15;17) translocation that generates the fusion protein promyelocytic leukemia-retinoic acid receptor α (PML-RARA) in nearly all cases. Multiple prior mouse models of APL constitutively express PML-RARA from a variety of non-Pml loci. Typically, all animals develop a myeloproliferative disease, followed by leukemia in a subset of animals after a long latent period. In contrast, human APL is not associated with an antecedent stage of myeloproliferation. To address this discrepancy, we have generated a system whereby PML-RARA expression is somatically acquired from the mouse Pml locus in the context of Pml haploinsufficiency. We found that physiologic PML-RARA expression was sufficient to direct a hematopoietic progenitor self-renewal program in vitro and in vivo. However, this expansion was not associated with evidence of myeloproliferation, more accurately reflecting the clinical presentation of human APL. Thus, at physiologic doses, PML-RARA primarily acts to increase hematopoietic progenitor self-renewal, expanding a population of cells that are susceptible to acquiring secondary mutations that cause progression to leukemia. This mouse model provides a platform for more accurately dissecting the early events in APL pathogenesis.
Collapse
Affiliation(s)
- John S Welch
- Section of Stem Cell Biology, Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63119, USA
| | | | | |
Collapse
|
43
|
The role of PML in the control of apoptotic cell fate: a new key player at ER-mitochondria sites. Cell Death Differ 2011; 18:1450-6. [PMID: 21475307 DOI: 10.1038/cdd.2011.31] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The development of malignant tumors results from deregulated proliferation or an inability of cells to undergo apoptotic cell death. Experimental works of the past decade have highlighted the importance of calcium (Ca(2+)) in the regulation of apoptosis. Several studies indicate that the Ca(2+) content of the endoplasmic reticulum (ER) determines the cell's sensitivity to apoptotic stress and perturbation of ER Ca(2+) homeostasis appears to be a key component in the development of several pathological situations. Sensitivity to apoptosis depends on the ability of cells to transfer Ca(2+) from the ER to the mitochondria. The physical platform for the interplay between the ER and mitochondria is a domain of the ER called the mitochondria-associated membranes (MAMs). The disruption of these contact sites has profound consequences for cellular function, such as imbalances of intracellular Ca(2+) signaling, cellular stress, and disrupted apoptosis progression. The promyelocytic leukemia (PML) protein has been previously recognized as a critical and essential regulator of multiple apoptotic response. Nevertheless, how PML would exert such broad and fundamental role in apoptosis remained for long time a mystery. In this review, we will discuss how recent results demonstrate that the elusive mechanism whereby the PML tumor suppressor exerts its essential role in apoptosis triggered by Ca(2+)-dependent stimuli can be attributed to its unexpected and fundamental role at MAMs in the control of the functional cross-talk between ER and mitochondria.
Collapse
|
44
|
The self-association coiled-coil domain of PML is sufficient for the oncogenic conversion of the retinoic acid receptor (RAR) alpha. Leukemia 2011; 25:814-20. [DOI: 10.1038/leu.2011.18] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
45
|
Scaglioni PP, Cai LF, Majid SM, Yung TM, Socci ND, Kogan SC, Kopelovich L, Pandolfi PP. Treatment with 5-azacytidine accelerates acute promyelocytic leukemia leukemogenesis in a transgenic mouse model. Genes Cancer 2011; 2:160-5. [PMID: 21779489 PMCID: PMC3111249 DOI: 10.1177/1947601911410300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 04/20/2011] [Accepted: 04/20/2011] [Indexed: 11/17/2022] Open
Abstract
A key oncogenic force in acute promyelocytic leukemia (APL) is the ability of the promyelocytic leukemia-retinoic acid receptor α (PML-RARA) oncoprotein to recruit transcriptional repressors and DNA methyltransferases at retinoic acid-responsive elements. Pharmacological doses of retinoic acid relieve transcriptional repression inducing terminal differentiation/apoptosis of the leukemic blasts. APL blasts often harbor additional recurrent chromosomal abnormalities, and significantly, APL prevalence is increased in Latino populations. These observations suggest that multiple genetic and environmental/dietary factors are likely implicated in APL. We tested whether dietary or targeted chemopreventive strategies relieving PML-RARA transcriptional repression would be effective in a transgenic mouse model. Surprisingly, we found that 1) treatment with a demethylating agent, 5-azacytidine, results in a striking acceleration of APL; 2) a high fat, low folate/choline-containing diet resulted in a substantial but nonsignificant APL acceleration; and 3) all-trans retinoic acid (ATRA) is ineffective in preventing leukemia and results in ATRA-resistant APL. Our findings have important clinical implications because ATRA is a drug of choice for APL treatment and indicate that global demethylation, whether through dietary manipulations or through the use of a pharmacologic agent such as 5-azacytidine, may have unintended and detrimental consequences in chemopreventive regimens.
Collapse
Affiliation(s)
- Pier Paolo Scaglioni
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Current address: Division of Hematology Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lu Fan Cai
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Samia M. Majid
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Thomas M. Yung
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Nicholas D. Socci
- Computational Biology Center, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Scott C. Kogan
- Department of Laboratory Medicine and Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Levy Kopelovich
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Pier Paolo Pandolfi
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Current address: Cancer Genetics Program, Beth Israel Deaconess Cancer Center, and Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Rara haploinsufficiency modestly influences the phenotype of acute promyelocytic leukemia in mice. Blood 2010; 117:2460-8. [PMID: 21190992 DOI: 10.1182/blood-2010-08-300087] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RARA (retinoic acid receptor alpha) haploinsufficiency is an invariable consequence of t(15;17)(q22;q21) translocations in acute promyelocytic leukemia (APL). Retinoids and RARA activity have been implicated in hematopoietic self-renewal and neutrophil maturation. We and others therefore predicted that RARA haploinsufficiency would contribute to APL pathogenesis. To test this hypothesis, we crossed Rara(+/-) mice with mice expressing PML (promyelocytic leukemia)-RARA from the cathepsin G locus (mCG-PR). We found that Rara haploinsufficiency cooperated with PML-RARA, but only modestly influenced the preleukemic and leukemic phenotype. Bone marrow from mCG-PR(+/-) × Rara(+/-) mice had decreased numbers of mature myeloid cells, increased ex vivo myeloid cell proliferation, and increased competitive advantage after transplantation. Rara haploinsufficiency did not alter mCG-PR-dependent leukemic latency or penetrance, but did influence the distribution of leukemic cells; leukemia in mCG-PR(+/-) × Rara(+/-) mice presented more commonly with low to normal white blood cell counts and with myeloid infiltration of lymph nodes. APL cells from these mice were responsive to all-trans retinoic acid and had virtually no differences in expression profiling compared with tumors arising in mCG-PR(+/-) × Rara(+/+) mice. These data show that Rara haploinsufficiency (like Pml haploinsufficiency and RARA-PML) can cooperate with PML-RARA to influence the pathogenesis of APL in mice, but that PML-RARA is the t(15;17) disease-initiating mutation.
Collapse
|
47
|
Negorev DG, Vladimirova OV, Kossenkov AV, Nikonova EV, Demarest RM, Capobianco AJ, Showe MK, Rauscher FJ, Showe LC, Maul GG. Sp100 as a potent tumor suppressor: accelerated senescence and rapid malignant transformation of human fibroblasts through modulation of an embryonic stem cell program. Cancer Res 2010; 70:9991-10001. [PMID: 21118961 PMCID: PMC3059726 DOI: 10.1158/0008-5472.can-10-1483] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Identifying the functions of proteins, which associate with specific subnuclear structures, is critical to understanding eukaryotic nuclear dynamics. Sp100 is a prototypical protein of ND10/PML nuclear bodies, which colocalizes with Daxx and the proto-oncogenic PML. Sp100 isoforms contain SAND, PHD, Bromo, and HMG domains and are highly sumoylated, all characteristics suggestive of a role in chromatin-mediated gene regulation. A role for Sp100 in oncogenesis has not been defined previously. Using selective Sp100 isoform-knockdown approaches, we show that normal human diploid fibroblasts with reduced Sp100 levels rapidly senesce. Subsequently, small rapidly dividing Sp100 minus cells emerge from the senescing fibroblasts and are found to be highly tumorigenic in nude mice. The derivation of these tumorigenic cells from the parental fibroblasts is confirmed by microsatellite analysis. The small rapidly dividing Sp100 minus cells now also lack ND10/PML bodies, and exhibit genomic instability and p53 cytoplasmic sequestration. They have also activated MYC, RAS, and TERT pathways and express mesenchymal to epithelial transdifferentiation (MET) markers. Reintroduction of expression of only the Sp100A isoform is sufficient to maintain senescence and to inhibit emergence of the highly tumorigenic cells. Global transcriptome studies, quantitative PCR, and protein studies, as well as immunolocalization studies during the course of the transformation, reveal that a transient expression of stem cell markers precedes the malignant transformation. These results identify a role for Sp100 as a tumor suppressor in addition to its role in maintaining ND10/PML bodies and in the epigenetic regulation of gene expression.
Collapse
MESH Headings
- Animals
- Antigens, Nuclear/genetics
- Antigens, Nuclear/metabolism
- Autoantigens/genetics
- Autoantigens/metabolism
- Blotting, Western
- Cell Transformation, Neoplastic/genetics
- Cells, Cultured
- Cellular Senescence/genetics
- Embryonic Stem Cells/metabolism
- Epithelial-Mesenchymal Transition/genetics
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Gene Expression Profiling
- HEK293 Cells
- Humans
- Male
- Mice
- Mice, Nude
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Nuclear Proteins/metabolism
- Oligonucleotide Array Sequence Analysis
- Promyelocytic Leukemia Protein
- Proto-Oncogene Proteins c-myc/metabolism
- RNA Interference
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription Factors/metabolism
- Transplantation, Heterologous
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- ras Proteins/metabolism
Collapse
Affiliation(s)
| | | | | | | | | | | | - Michael K. Showe
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
| | | | - Louise C. Showe
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
| | - Gerd G. Maul
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
| |
Collapse
|
48
|
BCOR as a novel fusion partner of retinoic acid receptor alpha in a t(X;17)(p11;q12) variant of acute promyelocytic leukemia. Blood 2010; 116:4274-83. [DOI: 10.1182/blood-2010-01-264432] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abstract
The majority of acute promyelocytic leukemia (APL) cases are characterized by the presence of a promyelocytic leukemia–retinoic acid receptor alpha(RARA) fusion gene. In a small subset, RARA is fused to a different partner, usually involved in regulating cell growth and differentiation. Here, we identified a novel RARA fusion transcript, BCOR-RARA, in a t(X;17)(p11;q12) variant of APL with unique morphologic features, including rectangular and round cytoplasmic inclusion bodies. Although the patient was clinically responsive to all-trans retinoic acid, several relapses occurred with standard chemotherapy and all-trans retinoic acid. BCOR is a transcriptional corepressor through the proto-oncoprotein, BCL6, recruiting histone deacetylases and polycomb repressive complex 1 components. BCOR-RARA was found to possess common features with other RARA fusion proteins. These included: (1) the same break point in RARA cDNA; (2) self-association; (3) retinoid X receptor alpha is necessary for BCOR-RARA to associate with the RARA responsive element; (4) action in a dominant-negative manner on RARA transcriptional activation; and (5) aberrant subcellular relocalization. It should be noted that there was no intact BCOR found in the 45,-Y,t(X;17)(p11;q12) APL cells because they featured only a rearranged X chromosome. These results highlight essential features of pathogenesis in APL in more detail. BCOR appears to be involved not only in human congenital diseases, but also in a human cancer.
Collapse
|
49
|
Reineke EL, Liu Y, Kao HY. Promyelocytic leukemia protein controls cell migration in response to hydrogen peroxide and insulin-like growth factor-1. J Biol Chem 2010; 285:9485-9492. [PMID: 20100838 PMCID: PMC2843199 DOI: 10.1074/jbc.m109.063362] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Revised: 01/19/2010] [Indexed: 01/14/2023] Open
Abstract
Promyelocytic leukemia protein (PML) was originally identified as part of a chromosomal translocation that contributes to the development of acute promyelocytic leukemia (APL). Since its discovery, PML has been found to play diverse roles in different cellular processes. Notably, PML has anti-proliferative and pro-apoptotic activity that supports its role as a tumor suppressor. We have previously shown that the peptidyl-prolyl isomerase Pin1 is able to affect cell proliferation and hydrogen peroxide (H(2)O(2))-mediated cell death through modulation of the steady-state levels of PML. We have extended these studies to show that the interaction between PML and Pin1 is targeted by multiple extracellular signals in the cell. We show that H(2)O(2) up-regulates and IGF-1 down-regulates PML expression in a Pin1-dependent manner. Interestingly, we found that H(2)O(2)- and IGF-1-mediated alteration in PML accumulation regulate MDA-MB-231 cell migration. Furthermore, we show that the control of cell migration by PML, and thus H(2)O(2) and IGF-1, results from PML-dependent decreased expression of integrin beta1 (ITGB1). Knockdown of Pin1 leads to decreased cell migration, lower levels of ITGB1 expression and resistance to IGF-1- and H(2)O(2)-induced changes in cell migration and ITGB1 expression. Taken together, our work identifies PML as a common target for H(2)O(2) and IGF-1 and supports a novel tumor suppressive role for PML in controlling cell migration through the expression of ITGB1.
Collapse
Affiliation(s)
- Erin L Reineke
- Department of Biochemistry, School of Medicine, Case Western Reserve University, the Case Comprehensive Cancer Center, and University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Yu Liu
- Department of Biochemistry, School of Medicine, Case Western Reserve University, the Case Comprehensive Cancer Center, and University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Hung-Ying Kao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, the Case Comprehensive Cancer Center, and University Hospitals of Cleveland, Cleveland, Ohio 44106.
| |
Collapse
|
50
|
Sharma P, Murillas R, Zhang H, Kuehn MR. N4BP1 is a newly identified nucleolar protein that undergoes SUMO-regulated polyubiquitylation and proteasomal turnover at promyelocytic leukemia nuclear bodies. J Cell Sci 2010; 123:1227-34. [PMID: 20233849 DOI: 10.1242/jcs.060160] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
A number of proteins can be conjugated with both ubiquitin and the small ubiquitin-related modifier (SUMO), with crosstalk between these two post-translational modifications serving to regulate protein function and stability. We previously identified N4BP1 as a substrate for monoubiquitylation by the E3 ubiquitin ligase Nedd4. Here, we describe Nedd4-mediated polyubiquitylation and proteasomal degradation of N4BP1. In addition, we show that N4BP1 can be conjugated with SUMO1 and that this abrogates N4BP1 ubiquitylation. Consistent with this, endogenous N4BP1 is stabilized in primary embryonic fibroblasts from mutants of the desumoylating enzyme SENP1, which show increased steady-state sumoylation levels. We have localized endogenous N4BP1 predominantly to the nucleolus in primary cells. However, a small fraction is found at promyelocytic leukemia (PML) nuclear bodies (NBs). In cells deficient for SENP1 or in wild-type cells treated with the proteasome inhibitor MG132, there is considerable accumulation of N4BP1 at PML NBs. These findings suggest a dynamic interaction between subnuclear compartments, and a role for post-translational modification by ubiquitin and SUMO in the regulation of nucleolar protein turnover.
Collapse
Affiliation(s)
- Prashant Sharma
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, National Cancer Institute, NCI-Frederick, Frederick, MD 21702, USA
| | | | | | | |
Collapse
|