1
|
Bercier P, Wang QQ, Zang N, Zhang J, Yang C, Maimaitiyiming Y, Abou-Ghali M, Berthier C, Wu C, Niwa-Kawakita M, Dirami T, Geoffroy MC, Ferhi O, Quentin S, Benhenda S, Ogra Y, Gueroui Z, Zhou C, Naranmandura H, de Thé H, Lallemand-Breitenbach V. Structural Basis of PML-RARA Oncoprotein Targeting by Arsenic Unravels a Cysteine Rheostat Controlling PML Body Assembly and Function. Cancer Discov 2023; 13:2548-2565. [PMID: 37655965 PMCID: PMC10714139 DOI: 10.1158/2159-8290.cd-23-0453] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 07/31/2023] [Accepted: 08/30/2023] [Indexed: 09/02/2023]
Abstract
PML nuclear bodies (NB) are disrupted in PML-RARA-driven acute promyelocytic leukemia (APL). Arsenic trioxide (ATO) cures 70% of patients with APL, driving PML-RARA degradation and NB reformation. In non-APL cells, arsenic binding onto PML also amplifies NB formation. Yet, the actual molecular mechanism(s) involved remain(s) elusive. Here, we establish that PML NBs display some features of liquid-liquid phase separation and that ATO induces a gel-like transition. PML B-box-2 structure reveals an alpha helix driving B2 trimerization and positioning a cysteine trio to form an ideal arsenic-binding pocket. Altering either of the latter impedes ATO-driven NB assembly, PML sumoylation, and PML-RARA degradation, mechanistically explaining clinical ATO resistance. This B2 trimer and the C213 trio create an oxidation-sensitive rheostat that controls PML NB assembly dynamics and downstream signaling in both basal state and during stress response. These findings identify the structural basis for arsenic targeting of PML that could pave the way to novel cancer drugs. SIGNIFICANCE Arsenic curative effects in APL rely on PML targeting. We report a PML B-box-2 structure that drives trimer assembly, positioning a cysteine trio to form an arsenic-binding pocket, which is disrupted in resistant patients. Identification of this ROS-sensitive triad controlling PML dynamics and functions could yield novel drugs. See related commentary by Salomoni, p. 2505. This article is featured in Selected Articles from This Issue, p. 2489.
Collapse
Affiliation(s)
- Pierre Bercier
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| | - Qian Qian Wang
- Department of Hematology of First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Public Health, School of Medicine and Department of Toxicology, Zhejiang University, Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ning Zang
- Public Health, School of Medicine and Department of Toxicology, Zhejiang University, Hangzhou, China
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Zhang
- Public Health, School of Medicine and Department of Toxicology, Zhejiang University, Hangzhou, China
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chang Yang
- Department of Hematology of First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Public Health, School of Medicine and Department of Toxicology, Zhejiang University, Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yasen Maimaitiyiming
- Department of Hematology of First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Public Health, School of Medicine and Department of Toxicology, Zhejiang University, Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Majdouline Abou-Ghali
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| | - Caroline Berthier
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Chengchen Wu
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| | - Michiko Niwa-Kawakita
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| | - Thassadite Dirami
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| | - Marie-Claude Geoffroy
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| | - Omar Ferhi
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| | - Samuel Quentin
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| | - Shirine Benhenda
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| | - Yasumitsu Ogra
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Zoher Gueroui
- Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France
| | - Chun Zhou
- Public Health, School of Medicine and Department of Toxicology, Zhejiang University, Hangzhou, China
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hua Naranmandura
- Department of Hematology of First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Public Health, School of Medicine and Department of Toxicology, Zhejiang University, Hangzhou, China
| | - Hugues de Thé
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
- Hematology Laboratory, Hôpital St Louis, AP/HP, Paris, France
| | - Valérie Lallemand-Breitenbach
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- GenCellDis, Inserm U944, CNRS UMR7212, Université Paris Cité, Paris, France
| |
Collapse
|
2
|
Bregnard TA, Fairchild D, Erlandsen H, Semenova IV, Szczepaniak R, Ahmed A, Weller SK, Korzhnev DM, Bezsonova I. Conformational exchange at a C 2H 2 zinc-binding site facilitates redox sensing by the PML protein. Structure 2023; 31:1086-1099.e6. [PMID: 37473756 PMCID: PMC10528520 DOI: 10.1016/j.str.2023.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/12/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023]
Abstract
The promyelocytic leukemia protein, PML, plays a vital role in the cellular response to oxidative stress; however, the molecular mechanism of its action remains poorly understood. Here, we identify redox-sensitive sites of PML. A molecule of PML is cysteine-rich and contains three zinc-binding domains including RING, B-box1, and B-box2. Using in vitro assays, we have compared the sensitivity of the isolated RING and B-box1 domains and shown that B-box1 is more sensitive to oxidation. NMR studies of PML dynamics showed that one of the Zn-coordination sites within the B-box1 undergoes significant conformational exchange, revealing a hotspot for exposure of reactive cysteines. In agreement with the in vitro data, enhancement of the B-box1 Zn-coordination dynamics led to more efficient recruitment of PML into PML nuclear bodies in cells. Overall, our results suggest that the increased sensitivity of B-box1 to oxidative stress makes this domain an important redox-sensing component of PML.
Collapse
Affiliation(s)
- Thomas A Bregnard
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, CT 06032, USA
| | - Daniel Fairchild
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, CT 06032, USA
| | - Heidi Erlandsen
- Center for Open Research Resources & Equipment, UCONN, Storrs, CT 06269, USA
| | - Irina V Semenova
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, CT 06032, USA
| | - Renata Szczepaniak
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, CT 06032, USA
| | - Affrin Ahmed
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, CT 06032, USA
| | - Sandra K Weller
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, CT 06032, USA
| | - Dmitry M Korzhnev
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, CT 06032, USA
| | - Irina Bezsonova
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, CT 06032, USA.
| |
Collapse
|
3
|
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
|
4
|
Liu Z, Qin Z, Liu Y, Xia X, He L, Chen N, Hu X, Peng X. Liquid‒liquid phase separation: roles and implications in future cancer treatment. Int J Biol Sci 2023; 19:4139-4156. [PMID: 37705755 PMCID: PMC10496506 DOI: 10.7150/ijbs.81521] [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: 12/04/2022] [Accepted: 07/23/2023] [Indexed: 09/15/2023] Open
Abstract
Liquid‒liquid phase separation (LLPS) is a phenomenon driven by weak interactions between biomolecules, such as proteins and nucleic acids, that leads to the formation of distinct liquid-like condensates. Through LLPS, membraneless condensates are formed, selectively concentrating specific proteins while excluding other molecules to maintain normal cellular functions. Emerging evidence shows that cancer-related mutations cause aberrant condensate assembly, resulting in disrupted signal transduction, impaired DNA repair, and abnormal chromatin organization and eventually contributing to tumorigenesis. The objective of this review is to summarize recent advancements in understanding the potential implications of LLPS in the contexts of cancer progression and therapeutic interventions. By interfering with LLPS, it may be possible to restore normal cellular processes and inhibit tumor progression. The underlying mechanisms and potential drug targets associated with LLPS in cancer are discussed, shedding light on promising opportunities for novel therapeutic interventions.
Collapse
Affiliation(s)
- Zheran Liu
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zijian Qin
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yingtong Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu 610041, Sichuan, China
| | - Xi Xia
- Shanghai ETERN Biopharma Co., Ltd., Shanghai, China
| | - Ling He
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Na Chen
- School of Pharmacy, Chengdu Medical College, Xindu Avenue No 783, Chengdu, 610500, Sichuan Province, China
| | - Xiaolin Hu
- West China School of Nursing, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
5
|
Exploration of nuclear body-enhanced sumoylation reveals that PML represses 2-cell features of embryonic stem cells. Nat Commun 2022; 13:5726. [PMID: 36175410 PMCID: PMC9522831 DOI: 10.1038/s41467-022-33147-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 09/05/2022] [Indexed: 01/12/2023] Open
Abstract
Membrane-less organelles are condensates formed by phase separation whose functions often remain enigmatic. Upon oxidative stress, PML scaffolds Nuclear Bodies (NBs) to regulate senescence or metabolic adaptation. PML NBs recruit many partner proteins, but the actual biochemical mechanism underlying their pleiotropic functions remains elusive. Similarly, PML role in embryonic stem cell (ESC) and retro-element biology is unsettled. Here we demonstrate that PML is essential for oxidative stress-driven partner SUMO2/3 conjugation in mouse ESCs (mESCs) or leukemia, a process often followed by their poly-ubiquitination and degradation. Functionally, PML is required for stress responses in mESCs. Differential proteomics unravel the KAP1 complex as a PML NB-dependent SUMO2-target in arsenic-treated APL mice or mESCs. PML-driven KAP1 sumoylation enables activation of this key epigenetic repressor implicated in retro-element silencing. Accordingly, Pml-/- mESCs re-express transposable elements and display 2-Cell-Like features, the latter enforced by PML-controlled SUMO2-conjugation of DPPA2. Thus, PML orchestrates mESC state by coordinating SUMO2-conjugation of different transcriptional regulators, raising new hypotheses about PML roles in cancer.
Collapse
|
6
|
Preethi S, Arthiga K, Patil AB, Spandana A, Jain V. Review on NAD(P)H dehydrogenase quinone 1 (NQO1) pathway. Mol Biol Rep 2022; 49:8907-8924. [PMID: 35347544 DOI: 10.1007/s11033-022-07369-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/11/2022] [Indexed: 12/14/2022]
Abstract
NQO1 is an enzyme present in humans which is encoded by NQO1 gene. It is a protective antioxidant agent, versatile cytoprotective agent and regulates the oxidative stresses of chromatin binding proteins for DNA damage in cancer cells. The oxidization of cellular pyridine nucleotides causes structural alterations to NQO1 and changes in its capacity to binding of proteins. A strategy based on NQO1 to have protective effect against cancer was developed by organic components to enhance NQO1 expression. The quinone derivative compounds like mitomycin C, RH1, E09 (Apaziquone) and β-lapachone causes cell death by NQO1 reduction of two electrons. It was also known to be overexpressed in various tumor cells of breast, lung, cervix, pancreas and colon when it was compared with normal cells in humans. The mechanism of NQO1 by the reduction of FAD by NADPH to form FADH2 is by two ways to inhibit cancer cell development such as suppression of carcinogenic metabolic activation and prevention of carcinogen formation. The NQO1 exhibit suppression of chemical-mediated carcinogenesis by various properties of NQO1 which includes, detoxification of quinone scavenger of superoxide anion radical, antioxidant enzyme, protein stabilizer. This review outlines the NQO1 structure, mechanism of action to inhibit the cancer cell, functions of NQO1 against oxidative stress, drugs acting on NQO1 pathways, clinical significance.
Collapse
Affiliation(s)
- S Preethi
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India
| | - K Arthiga
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India
| | - Amit B Patil
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India
| | - Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka, 570015, India.
| |
Collapse
|
7
|
Wu HC, Rérolle D, Berthier C, Hleihel R, Sakamoto T, Quentin S, Benhenda S, Morganti C, Wu C, Conte L, Rimsky S, Sebert M, Clappier E, Souquere S, Gachet S, Soulier J, Durand S, Trowbridge JJ, Bénit P, Rustin P, El Hajj H, Raffoux E, Ades L, Itzykson R, Dombret H, Fenaux P, Espeli O, Kroemer G, Brunetti L, Mak TW, Lallemand-Breitenbach V, Bazarbachi A, Falini B, Ito K, Martelli MP, de Thé H. Actinomycin D Targets NPM1c-Primed Mitochondria to Restore PML-Driven Senescence in AML Therapy. Cancer Discov 2021; 11:3198-3213. [PMID: 34301789 PMCID: PMC7612574 DOI: 10.1158/2159-8290.cd-21-0177] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/07/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) pathogenesis often involves a mutation in the NPM1 nucleolar chaperone, but the bases for its transforming properties and overall association with favorable therapeutic responses remain incompletely understood. Here we demonstrate that an oncogenic mutant form of NPM1 (NPM1c) impairs mitochondrial function. NPM1c also hampers formation of promyelocytic leukemia (PML) nuclear bodies (NB), which are regulators of mitochondrial fitness and key senescence effectors. Actinomycin D (ActD), an antibiotic with unambiguous clinical efficacy in relapsed/refractory NPM1c-AMLs, targets these primed mitochondria, releasing mitochondrial DNA, activating cyclic GMP-AMP synthase signaling, and boosting reactive oxygen species (ROS) production. The latter restore PML NB formation to drive TP53 activation and senescence of NPM1c-AML cells. In several models, dual targeting of mitochondria by venetoclax and ActD synergized to clear AML and prolong survival through targeting of PML. Our studies reveal an unexpected role for mitochondria downstream of NPM1c and implicate a mitochondrial/ROS/PML/TP53 senescence pathway as an effector of ActD-based therapies. SIGNIFICANCE ActD induces complete remissions in NPM1-mutant AMLs. We found that NPM1c affects mitochondrial biogenesis and PML NBs. ActD targets mitochondria, yielding ROS which enforce PML NB biogenesis and restore senescence. Dual targeting of mitochondria with ActD and venetoclax sharply potentiates their anti-AML activities in vivo. This article is highlighted in the In This Issue feature, p. 2945.
Collapse
Affiliation(s)
- Hsin-Chieh Wu
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Domitille Rérolle
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Caroline Berthier
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Rita Hleihel
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Internal Medicine and Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
- Department of Experimental Pathology, Microbiology and Immunology, American University of Beirut, Beirut, Lebanon
| | - Takashi Sakamoto
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Samuel Quentin
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Shirine Benhenda
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Claudia Morganti
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research and Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Chengchen Wu
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Lidio Conte
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli, ” Napoli, Italy
| | - Sylvie Rimsky
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
| | - Marie Sebert
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Emmanuelle Clappier
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Sylvie Souquere
- Institut Gustave Roussy, Cell Biology and Metabolomics Platforms, INSERM UMS 3655, Villejuif, France
| | - Stéphanie Gachet
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Jean Soulier
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Sylvère Durand
- Institut Gustave Roussy, Cell Biology and Metabolomics Platforms, INSERM UMS 3655, Villejuif, France
| | | | - Paule Bénit
- INSERM, U1141 Hôpital Robert Debré, Paris France
| | | | - Hiba El Hajj
- Department of Experimental Pathology, Microbiology and Immunology, American University of Beirut, Beirut, Lebanon
| | - Emmanuel Raffoux
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Lionel Ades
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Raphael Itzykson
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Hervé Dombret
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Pierre Fenaux
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Olivier Espeli
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
| | - Guido Kroemer
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli, ” Napoli, Italy
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Lorenzo Brunetti
- Hematology, Department of Medicine and surgery, University of Perugia, Perugia, Italy
| | - Tak W. Mak
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Valérie Lallemand-Breitenbach
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Ali Bazarbachi
- Department of Internal Medicine and Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Brunangelo Falini
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research and Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, New York
| | | | - Hugues de Thé
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| |
Collapse
|
8
|
Yi J, Peng J, Yang W, Zhu G, Ren J, Li D, Zheng H. Picornavirus 3C - a protease ensuring virus replication and subverting host responses. J Cell Sci 2021; 134:134/5/jcs253237. [PMID: 33692152 DOI: 10.1242/jcs.253237] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The protease 3C is encoded by all known picornaviruses, and the structural features related to its protease and RNA-binding activities are conserved; these contribute to the cleavage of viral polyproteins and the assembly of the viral RNA replication complex during virus replication. Furthermore, 3C performs functions in the host cell through its interaction with host proteins. For instance, 3C has been shown to selectively 'hijack' host factors involved in gene expression, promoting picornavirus replication, and to inactivate key factors in innate immunity signaling pathways, inhibiting the production of interferon and inflammatory cytokines. Importantly, 3C maintains virus infection by subtly subverting host cell death and modifying critical molecules in host organelles. This Review focuses on the molecular mechanisms through which 3C mediates physiological processes involved in virus-host interaction, thus highlighting the picornavirus-mediated pathogenesis caused by 3C.
Collapse
Affiliation(s)
- Jiamin Yi
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Jiangling Peng
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Wenping Yang
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Guoqiang Zhu
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Jingjing Ren
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Dan Li
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| |
Collapse
|
9
|
Li Y, Chandra TP, Song X, Nie L, Liu M, Yi J, Zheng X, Chu C, Yang J. H2S improves doxorubicin-induced myocardial fibrosis by inhibiting oxidative stress and apoptosis via Keap1-Nrf2. Technol Health Care 2021; 29:195-209. [PMID: 33682759 PMCID: PMC8150551 DOI: 10.3233/thc-218020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE We waimed to investigate whether H2S can relieve the myocardial fibrosis caused by doxorubicin through Keap1-Nrf2. METHODS Sprague-Dawley (SD) rats were randomly divided into four groups: normal control group (Control); DOX model group (DOX); H2S intervention model group (DOX+H2S); H2S control group (H2S). DOX and DOX+H2S group were injected with doxorubicin (3.0 mg/kg/time) intraperitoneally. Both of the Control group and H2S groups were given normal saline in equal volume, 2 weeks later, DOX+H2S and H2S group were controlled with NaHS (56 μmol/kg/d) through the abdominal cavity, while the Control and DOX group were injected with normal saline of the same dosage intraperitoneally. RESULTS Myocardial injury and myocardial cell apoptosis were significantly increased, the H2S content in myocardial tissue was remarkably down-regulated, the expression levels of MDA, Keap1, caspase-3, caspase-9, TNF-α, IL1β, MMPs and TIMP-1 in rat myocardial tissue was significantly up-regulated (P< 0.05), and the expression levels of GSH, NQO1, Bcl-2 were down-regulated compared with those of control group. The above results can be reversed by the DOX+H2S group. There is no statistically significant difference between the Control group and the H2S control group. CONCLUSIONS These results suggest that H2S can improve DOX-induced myocardial fibrosis in rats, and the keap1/Nrf2 signaling pathway, oxidative stress, inflammation, and apoptosis may be involved in the mechanism.
Collapse
Affiliation(s)
- Yaling Li
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Thakur Prakash Chandra
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Xiong Song
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Maojun Liu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Jiali Yi
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Xia Zheng
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
10
|
Alhazmi N, Pai CP, Albaqami A, Wang H, Zhao X, Chen M, Hu P, Guo S, Starost K, Hajihassani O, Miyagi M, Kao HY. The promyelocytic leukemia protein isoform PML1 is an oncoprotein and a direct target of the antioxidant sulforaphane (SFN). BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118707. [PMID: 32243901 DOI: 10.1016/j.bbamcr.2020.118707] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 03/18/2020] [Accepted: 03/21/2020] [Indexed: 01/30/2023]
Abstract
The gene encoding promyelocytic leukemia protein (PML) generates several spliced isoforms. Ectopic expression of PML1 promotes the proliferation of ERα-positive MCF-7 breast cancer (BC) cells, while a loss of PML by knockdown or overexpression of PML4 does the opposite. PML is an essential constituent of highly dynamic particles called PML nuclear bodies (NBs). PML NBs are heterogenous multiprotein subnuclear structures that are part of cellular stress sensing machinery. The antioxidant sulforaphane (SFN) inhibits the proliferation of BC cells and causes a redistribution of the subcellular localization of PML, a disruption of disulfide-bond linkages in nuclear PML-containing complexes, and a reduction in the number and size of PML NBs. Mechanistically, SFN modifies several cysteine residues, including C204, located in the RBCC domain of PML. PML is sumoylated and contains a Sumo-interacting motif, and a significant fraction of Sumo1 and Sumo2/3 co-localizes with PML NBs. Ectopic expression of the mutant C204A selectively inhibits the biogenesis of endogenous PML NBs but not PML-less Sumo1-, Sumo2/3, or Daxx-containing nuclear speckles. Importantly, PML1 (C204A) functions as a dominant-negative mutant over endogenous PML protein and promotes anti-proliferation activity. Together, we conclude that SFN elicits its cytotoxic activity in part by inactivating PML1's pro-tumorigenic activity.
Collapse
Affiliation(s)
- Nada Alhazmi
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Chun-Peng Pai
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Aljawharah Albaqami
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Han Wang
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Xuan Zhao
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Minyue Chen
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Po Hu
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Shuang Guo
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Kyle Starost
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Omid Hajihassani
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Masaru Miyagi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Hung-Ying Kao
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU), 10900 Euclid Avenue, Cleveland, OH 44106, USA; The Comprehensive Cancer Center of CWRU, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| |
Collapse
|
11
|
Shytaj IL, Lucic B, Forcato M, Penzo C, Billingsley J, Laketa V, Bosinger S, Stanic M, Gregoretti F, Antonelli L, Oliva G, Frese CK, Trifunovic A, Galy B, Eibl C, Silvestri G, Bicciato S, Savarino A, Lusic M. Alterations of redox and iron metabolism accompany the development of HIV latency. EMBO J 2020; 39:e102209. [PMID: 32157726 PMCID: PMC7196916 DOI: 10.15252/embj.2019102209] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 12/19/2022] Open
Abstract
HIV-1 persists in a latent form during antiretroviral therapy, mainly in CD4+ T cells, thus hampering efforts for a cure. HIV-1 infection is accompanied by metabolic alterations, such as oxidative stress, but the effect of cellular antioxidant responses on viral replication and latency is unknown. Here, we show that cells survive retroviral replication, both in vitro and in vivo in SIVmac-infected macaques, by upregulating antioxidant pathways and the intertwined iron import pathway. These changes are associated with remodeling of promyelocytic leukemia protein nuclear bodies (PML NBs), an important constituent of nuclear architecture and a marker of HIV-1 latency. We found that PML NBs are hyper-SUMOylated and that PML protein is degraded via the ubiquitin-proteasome pathway in productively infected cells, before latency establishment and after reactivation. Conversely, normal numbers of PML NBs were restored upon transition to latency or by decreasing oxidative stress or iron content. Our results highlight antioxidant and iron import pathways as determinants of HIV-1 latency and support their pharmacologic inhibition as tools to regulate PML stability and impair latency establishment.
Collapse
Affiliation(s)
- Iart Luca Shytaj
- Heidelberg University HospitalHeidelbergGermany
- German Center for Infection ResearchHeidelbergGermany
| | - Bojana Lucic
- Heidelberg University HospitalHeidelbergGermany
- German Center for Infection ResearchHeidelbergGermany
| | - Mattia Forcato
- Department of Life SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | | | - James Billingsley
- Division of Microbiology and ImmunologyYerkes National Primate Research CenterEmory UniversityAtlantaGAUSA
| | - Vibor Laketa
- Heidelberg University HospitalHeidelbergGermany
- German Center for Infection ResearchHeidelbergGermany
| | - Steven Bosinger
- Division of Microbiology and ImmunologyYerkes National Primate Research CenterEmory UniversityAtlantaGAUSA
- Department of Pathology and Laboratory MedicineEmory UniversityAtlantaGAUSA
| | - Mia Stanic
- Heidelberg University HospitalHeidelbergGermany
| | | | - Laura Antonelli
- Institute for High Performance Computing and NetworkingICAR‐CNRNaplesItaly
| | - Gennaro Oliva
- Institute for High Performance Computing and NetworkingICAR‐CNRNaplesItaly
| | | | | | - Bruno Galy
- Division of Virus‐Associated CarcinogenesisGerman Cancer Research CentreHeidelbergGermany
| | - Clarissa Eibl
- Leibniz‐Forschungsinstitut für Molekulare PharmakologieBerlinGermany
- Institute of BiologyCellular BiophysicsHumboldt Universität zu BerlinBerlinGermany
| | - Guido Silvestri
- Division of Microbiology and ImmunologyYerkes National Primate Research CenterEmory UniversityAtlantaGAUSA
- Department of Pathology and Laboratory MedicineEmory UniversityAtlantaGAUSA
| | - Silvio Bicciato
- Department of Life SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | | | - Marina Lusic
- Heidelberg University HospitalHeidelbergGermany
- German Center for Infection ResearchHeidelbergGermany
| |
Collapse
|
12
|
Chen YT, Ou Yang WT, Juang HH, Chen CL, Chen HW, Tsui KH, Chang YH, Tsai CH, Hsueh C, Liao WC. Proteomic characterization of arsenic and cadmium exposure in bladder cells. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34 Suppl 1:e8578. [PMID: 31499585 DOI: 10.1002/rcm.8578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 05/22/2023]
Abstract
RATIONALE Accumulating evidence has linked prolonged exposure to heavy metals to cancer occurrence in the urinary system. However, the specific biological mechanisms responsible for the association of heavy metals with the unusually high incidence of upper tract urothelial carcinoma in Taiwan are complex and incompletely understood. METHODS To elucidate the specific biological mechanism and identify molecular indicators of the unusually high association of upper tract urothelial carcinoma with heavy metal exposure, protein expression following the treatment of T24 human bladder carcinoma and RT4 human bladder papilloma cell line models with arsenic (As) and cadmium (Cd) was studied. Proteomic changes in these cell models were integrated with data from a human bladder cancer (BLCA) tissue proteome to identify possible protein indicators of heavy metal exposure. RESULTS After mass spectrometry based proteomic analysis and verification by Western blotting procedures, we identified 66 proteins that were up-regulated and 92 proteins that were down-regulated in RT4 cell extracts after treatment with As or Cd. Some 52 proteins were up-regulated and 136 proteins were down-regulated in T24 cell extracts after treatment with Cd. We further confirmed that down-expression of the PML (promyelocytic leukemia) protein was sustained for at least 75 days after exposure of bladder cells to As. Dysregulation of these cellular proteins by As was associated with three biological pathways. Immunohistochemical analyses of paraffin-embedded BLCA tissue slides confirmed that PML protein expression was decreased in BLCA tumor cells compared with adjacent noncancerous epithelial cells. CONCLUSIONS These data suggest that PML may play an important role in the pathogenesis of BLCA and may be an indicator of heavy metal exposure in bladder cells.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Molecular Medicine Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Wei-Ting Ou Yang
- Molecular Medicine Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Anatomy, College of Medicine, Chang Gung University, TaoYuan, Taiwan
| | - Chien-Lun Chen
- Department of Urology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiao-Wei Chen
- Molecular Medicine Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ying-Hsu Chang
- Division of Urology, Department of Surgery, LinKou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Han Tsai
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chuen Hsueh
- Molecular Medicine Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Pathology, Chang Gung Memorial Hospital, Linkou, Chang Gung University, College of Medicine, Taoyuan, Taiwan
| | - Wei-Chao Liao
- Molecular Medicine Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Otolaryngology - Head & Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Center for General Education, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
13
|
Briones-Herrera A, Eugenio-Pérez D, Reyes-Ocampo JG, Rivera-Mancía S, Pedraza-Chaverri J. New highlights on the health-improving effects of sulforaphane. Food Funct 2018; 9:2589-2606. [PMID: 29701207 DOI: 10.1039/c8fo00018b] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this paper, we review recent evidence about the beneficial effects of sulforaphane (SFN), which is the most studied member of isothiocyanates, on both in vivo and in vitro models of different diseases, mainly diabetes and cancer. The role of SFN on oxidative stress, inflammation, and metabolism is discussed, with emphasis on those nuclear factor E2-related factor 2 (Nrf2) pathway-mediated mechanisms. In the case of the anti-inflammatory effects of SFN, the point of convergence seems to be the downregulation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), with the consequent amelioration of other pathogenic processes such as hypertrophy and fibrosis. We emphasized that SFN shows opposite effects in normal and cancer cells at many levels; for instance, while in normal cells it has protective actions, in cancer cells it blocks the induction of factors related to the malignity of tumors, diminishes their development, and induces cell death. SFN is able to promote apoptosis in cancer cells by many mechanisms, the production of reactive oxygen species being one of the most relevant ones. Given its properties, SFN could be considered as a phytochemical at the forefront of natural medicine.
Collapse
Affiliation(s)
- Alfredo Briones-Herrera
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | | | | | | |
Collapse
|
14
|
Chen D, Feng C, Tian X, Zheng N, Wu Z. Promyelocytic Leukemia Restricts Enterovirus 71 Replication by Inhibiting Autophagy. Front Immunol 2018; 9:1268. [PMID: 29922292 PMCID: PMC5996053 DOI: 10.3389/fimmu.2018.01268] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 05/22/2018] [Indexed: 12/18/2022] Open
Abstract
The promyelocytic leukemia (PML) protein, also known as TRIM19, functions as a major organizer of PML nuclear bodies (NBs) in most mammalian cells and plays important roles in antiviral activities against both DNA and RNA viruses. In this study, we found that the downregulation of PML rendered HeLa cells more susceptible to infection by enterovirus 71 (EV71), and the overexpression of the PMLIII or PMLIV isoforms inhibited viral protein expression and resulted in viral titers that were 2–3 log units lower than those in the control. Using short interfering RNAs, the downregulation of either the PMLIII or PMLIV isoform increased both viral protein VP1 expression and viral production. The PML repression of EV71 replication was partially mediated by the inhibition of autophagy, and PML deficiency triggered autophagy. Furthermore, the EV71 infection resulted in a reduction in PML independent of the proteasome pathway. Instead, PML degradation was mediated by virus protease 3Cpro. In conclusion, PML contributes to a cellular antiviral effect by inhibiting autophagy, which is countered by a disruption of promyelocytic leukemia protein-nuclear bodies mediated by viral protease 3Cpro.
Collapse
Affiliation(s)
- Deyan Chen
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Chunhong Feng
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Xiaoyan Tian
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Nan Zheng
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China.,State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.,Medical School, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China.,State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.,Medical School, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
15
|
Burroughs AF, Eluhu S, Whalen D, Goodwin JS, Sakwe AM, Arinze IJ. PML-Nuclear Bodies Regulate the Stability of the Fusion Protein Dendra2-Nrf2 in the Nucleus. Cell Physiol Biochem 2018; 47:800-816. [PMID: 29807365 PMCID: PMC6503657 DOI: 10.1159/000490033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 04/11/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND/AIMS Nuclear factor erythroid 2-related factor 2 (Nrf2) is a basic leucine-zipper transcription factor essential for cellular responses to oxidative stress. Degradation of Nrf2 in the cytoplasm, mediated by Keap1-Cullin3/RING box1 (Cul3-Rbx1) E3 ubiquitin ligase and the proteasome, is considered the primary pathway controlling the cellular abundance of Nrf2. Although the nucleus has been implicated in the degradation of Nrf2, little information is available on how this compartment participates in degrading Nrf2. METHODS Here, we fused the photoconvertible fluorescent protein Dendra2 to Nrf2 and capitalized on the irreversible change in color (green to red) that occurs when Dendra2 undergoes photoconversion to study degradation of Dendra2-Nrf2 in single live cells. RESULTS Using this approach, we show that the half-life (t1/2) of Dendra2-Nrf2 in the whole cell, under homeostatic conditions, is 35 min. Inhibition of the proteasome with MG-132 or induction of oxidative stress with tert-butylhydroquinone (tBHQ) extended the half-life of Dendra2-Nrf2 by 6- and 28-fold, respectively. By inhibiting nuclear export using Leptomycin B, we provide direct evidence that degradation of Nrf2 also occurs in the nucleus and involves PML-NBs (Promyelocytic Leukemia-nuclear bodies). We further demonstrate that co-expression of Dendra2-Nrf2 and Crimson-PML-I lacking two PML-I sumoylation sites (K65R and K490R) changed the decay rate of Dendra2-Nrf2 in the nucleus and stabilized the nuclear derived Nrf2 levels in whole cells. CONCLUSION Altogether, our findings provide direct evidence for degradation of Nrf2 in the nucleus and suggest that modification of Nrf2 in PML nuclear bodies contributes to its degradation in intact cells.
Collapse
Affiliation(s)
| | - Sylvia Eluhu
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Diva Whalen
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - J. Shawn Goodwin
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Amos M. Sakwe
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Ifeanyi J. Arinze
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
16
|
Hsu KS, Kao HY. PML: Regulation and multifaceted function beyond tumor suppression. Cell Biosci 2018; 8:5. [PMID: 29416846 PMCID: PMC5785837 DOI: 10.1186/s13578-018-0204-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 01/12/2018] [Indexed: 01/15/2023] Open
Abstract
Promyelocytic leukemia protein (PML) was originally identified as a fusion partner of retinoic acid receptor alpha in acute promyelocytic leukemia patients with the (15;17) chromosomal translocation, giving rise to PML–RARα and RARα–PML fusion proteins. A body of evidence indicated that PML possesses tumor suppressing activity by regulating apoptosis, cell cycle, senescence and DNA damage responses. PML is enriched in discrete nuclear substructures in mammalian cells with 0.2–1 μm diameter in size, referred to as alternately Kremer bodies, nuclear domain 10, PML oncogenic domains or PML nuclear bodies (NBs). Dysregulation of PML NB formation results in altered transcriptional regulation, protein modification, apoptosis and cellular senescence. In addition to PML NBs, PML is also present in nucleoplasm and cytoplasmic compartments, including the endoplasmic reticulum and mitochondria-associated membranes. The role of PML in tumor suppression has been extensively studied but increasing evidence indicates that PML also plays versatile roles in stem cell renewal, metabolism, inflammatory responses, neural function, mammary development and angiogenesis. In this review, we will briefly describe the known PML regulation and function and include new findings.
Collapse
Affiliation(s)
- Kuo-Sheng Hsu
- 1Department of Biochemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA.,Present Address: Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Hung-Ying Kao
- 1Department of Biochemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA.,The Comprehensive Cancer Center of Case Western Reserve University and University Hospitals of Cleveland, Cleveland, OH 44106 USA
| |
Collapse
|
17
|
PML nuclear bodies, membrane-less domains acting as ROS sensors? Semin Cell Dev Biol 2017; 80:29-34. [PMID: 29157919 DOI: 10.1016/j.semcdb.2017.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 12/23/2022]
Abstract
PML Nuclear bodies (PML NBs) are spherical domains associated with a broad range of activities upon stress responses such as apoptosis, senescence DNA repair, epigenetic control, as well as control of oncogenesis. These bodies are considered as privileged sites for post-translational modifications, where sumoylation plays a key role. Here we summarize recent in vitro and in vivo findings on the link between PML NBs and ROS, in particular PML contributions to oxidative stress response. We discuss how it may regulate switch from cell protection against stress to cell arrest/cell death.
Collapse
|
18
|
Niwa-Kawakita M, Ferhi O, Soilihi H, Le Bras M, Lallemand-Breitenbach V, de Thé H. PML is a ROS sensor activating p53 upon oxidative stress. J Exp Med 2017; 214:3197-3206. [PMID: 28931625 PMCID: PMC5679165 DOI: 10.1084/jem.20160301] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/21/2017] [Accepted: 08/09/2017] [Indexed: 12/30/2022] Open
Abstract
Promyelocytic leukemia (PML) nuclear bodies modulate several processes, including senescence or apoptosis. Niwa-Kawakita et al. demonstrate that PML regulates reactive oxygen species (ROS) homeostasis in vivo by coupling ROS to p53 signaling to enforce basal ROS protection and mediate their acute toxicity. Promyelocytic leukemia (PML) nuclear bodies (NBs) recruit partner proteins, including p53 and its regulators, thereby controlling their abundance or function. Investigating arsenic sensitivity of acute promyelocytic leukemia, we proposed that PML oxidation promotes NB biogenesis. However, physiological links between PML and oxidative stress response in vivo remain unexplored. Here, we identify PML as a reactive oxygen species (ROS) sensor. Pml−/− cells accumulate ROS, whereas PML expression decreases ROS levels. Unexpectedly, Pml−/− embryos survive acute glutathione depletion. Moreover, Pml−/− animals are resistant to acetaminophen hepatotoxicity or fasting-induced steatosis. Molecularly, Pml−/− animals fail to properly activate oxidative stress–responsive p53 targets, whereas the NRF2 response is amplified and accelerated. Finally, in an oxidative stress–prone background, Pml−/− animals display a longevity phenotype, likely reflecting decreased basal p53 activation. Thus, similar to p53, PML exerts basal antioxidant properties but also drives oxidative stress–induced changes in cell survival/proliferation or metabolism in vivo. Through NB biogenesis, PML therefore couples ROS sensing to p53 responses, shedding a new light on the role of PML in senescence or stem cell biology.
Collapse
Affiliation(s)
- Michiko Niwa-Kawakita
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France.,Institut National de la Santé et de la Recherche Medicale (INSERM) UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Paris, France.,Centre National de la Recherche Scientifique (CNRS) UMR 7212, Paris, France
| | - Omar Ferhi
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France.,Institut National de la Santé et de la Recherche Medicale (INSERM) UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Paris, France.,Centre National de la Recherche Scientifique (CNRS) UMR 7212, Paris, France.,Collège de France, PSL Research University, Paris, France
| | - Hassane Soilihi
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France.,Institut National de la Santé et de la Recherche Medicale (INSERM) UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Paris, France.,Centre National de la Recherche Scientifique (CNRS) UMR 7212, Paris, France
| | - Morgane Le Bras
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France.,Institut National de la Santé et de la Recherche Medicale (INSERM) UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Paris, France.,Centre National de la Recherche Scientifique (CNRS) UMR 7212, Paris, France
| | - Valérie Lallemand-Breitenbach
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France.,Institut National de la Santé et de la Recherche Medicale (INSERM) UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Paris, France.,Centre National de la Recherche Scientifique (CNRS) UMR 7212, Paris, France.,Collège de France, PSL Research University, Paris, France
| | - Hugues de Thé
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, Paris, France .,Institut National de la Santé et de la Recherche Medicale (INSERM) UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Paris, France.,Centre National de la Recherche Scientifique (CNRS) UMR 7212, Paris, France.,Service de Biochimie, Assistance Publique Hôpitaux de Paris, Hôpital St. Louis, Paris, France.,Collège de France, PSL Research University, Paris, France
| |
Collapse
|
19
|
Tessier S, Martin-Martin N, de Thé H, Carracedo A, Lallemand-Breitenbach V. Promyelocytic Leukemia Protein, a Protein at the Crossroad of Oxidative Stress and Metabolism. Antioxid Redox Signal 2017; 26:432-444. [PMID: 27758112 DOI: 10.1089/ars.2016.6898] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Cellular metabolic activity impacts the production of reactive oxygen species (ROS), both positively through mitochondrial oxidative processes and negatively by promoting the production of reducing agents (including NADPH and reduced glutathione). A defined metabolic state in cancer cells is critical for cell growth and long-term self-renewal, and such state is intrinsically associated with redox balance. Promyelocytic leukemia protein (PML) regulates several biological processes, at least in part, through its ability to control the assembly of PML nuclear bodies (PML NBs). Recent Advances: PML is oxidation-prone, and oxidative stress promotes NB biogenesis. These nuclear subdomains recruit many nuclear proteins and regulate their SUMOylation and other post-translational modifications. Some of these cargos-such as p53, SIRT1, AKT, and mammalian target of rapamycin (mTOR)-are key regulators of cell fate. PML was also recently shown to regulate oxidation. CRITICAL ISSUES While it was long considered primarily as a tumor suppressor protein, PML-regulated metabolic switch uncovered that this protein could promote survival and/or stemness of some normal or cancer cells. In this study, we review the recent findings on this multifunctional protein. FUTURE DIRECTIONS Studying PML scaffolding functions as well as its fine role in the activation of p53 or fatty acid oxidation will bring new insights in how PML could bridge oxidative stress, senescence, cell death, and metabolism. Antioxid. Redox Signal. 26, 432-444.
Collapse
Affiliation(s)
- Sarah Tessier
- 1 Collège de France , Paris, France .,2 INSERM UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie , Paris, France .,3 CNRS UMR 7212 , Paris France .,4 Université Paris Diderot-Sorbonne Paris Cité , Paris, France
| | | | - Hugues de Thé
- 1 Collège de France , Paris, France .,2 INSERM UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie , Paris, France .,3 CNRS UMR 7212 , Paris France .,4 Université Paris Diderot-Sorbonne Paris Cité , Paris, France .,6 AP-HP, Service de Biochimie, Hôpital St. Louis , Paris, France
| | - Arkaitz Carracedo
- 5 CIC bioGUNE , Bizkaia Technology Part, Derio, Spain .,7 IKERBASQUE , Basque Foundation for Science, Bilbao, Spain .,8 Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU) , Bilbao, Spain
| | - Valérie Lallemand-Breitenbach
- 1 Collège de France , Paris, France .,2 INSERM UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie , Paris, France .,3 CNRS UMR 7212 , Paris France .,4 Université Paris Diderot-Sorbonne Paris Cité , Paris, France
| |
Collapse
|
20
|
Wu JM, Oraee A, Doonan BB, Pinto JT, Hsieh TC. Activation of NQO1 in NQO1*2 polymorphic human leukemic HL-60 cells by diet-derived sulforaphane. Exp Hematol Oncol 2016; 5:27. [PMID: 27625902 PMCID: PMC5020469 DOI: 10.1186/s40164-016-0056-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 08/30/2016] [Indexed: 02/06/2023] Open
Abstract
Background The NAD(P)H: quinone oxidoreductase (NQO1) confers protection against semiquinones and also elicits oxidative stress. The C609T polymorphism of the NQO1 gene, designated NQO1*2, significantly reduces its enzymatic activity due to rapid degradation of protein. Since down regulation of NQO1 mRNA expression correlates with increased susceptibility for developing different types of cancers, we investigated the link between leukemia and the NQO1*2 genotype by mining a web-based microarray dataset, ONCOMINE. Phytochemicals prevent DNA damage through activation of phase II detoxification enzymes including NQO1. Whether NQO1 expression/activity in leukemia cells that carry the labile NQO1*2 genotype can be induced by broccoli-derived phytochemical sulforaphane (SFN) is currently unknown. Methods and Results The ONCOMINE query showed that: (1) acute lymphoblastic leukemia and chronic myelogenous leukemia are associated with reduced NQO1 levels, and (2) under-expressed NQO1 was found in human HL-60 leukemia cell line containing the heterozygous NQO1*2 polymorphism. We examined induction of NQO1 activity/expression by SFN in HL-60 cells. A dose-dependent increase in NQO1 level/activity is accompanied by upregulation of the transcription factor, Nrf2, following 1–10 μM SFN treatment. Treatment with 25 µM SFN drastically reduced NQO1 levels, inhibited cell proliferation, caused sub-G1 cell arrest, and induced apoptosis, and a decrease in the levels of the transcription factor, nuclear factor-κB (NFκB). Conclusions Up to 10 μM of SFN increases NQO1 expression and suppresses HL-60 cell proliferation whereas ≥ 25 μM of SFN induces apoptosis in HL-60 cells. Further, SFN treatment restores NQO1 activity/levels in HL-60 cells expressing the NQO1*2 genotype.
Collapse
Affiliation(s)
- Joseph M Wu
- Room 147, Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, 15 Dana Road, Valhalla, NY 10595 USA
| | - Ardalan Oraee
- Room 147, Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, 15 Dana Road, Valhalla, NY 10595 USA
| | - Barbara B Doonan
- Room 147, Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, 15 Dana Road, Valhalla, NY 10595 USA
| | - John T Pinto
- Room 147, Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, 15 Dana Road, Valhalla, NY 10595 USA
| | - Tze-Chen Hsieh
- Room 147, Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, 15 Dana Road, Valhalla, NY 10595 USA
| |
Collapse
|
21
|
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
|
22
|
Sieprath T, Corne TDJ, Nooteboom M, Grootaert C, Rajkovic A, Buysschaert B, Robijns J, Broers JLV, Ramaekers FCS, Koopman WJH, Willems PHGM, De Vos WH. Sustained accumulation of prelamin A and depletion of lamin A/C both cause oxidative stress and mitochondrial dysfunction but induce different cell fates. Nucleus 2015; 6:236-46. [PMID: 25996284 PMCID: PMC4615646 DOI: 10.1080/19491034.2015.1050568] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The cell nucleus is structurally and functionally organized by lamins, intermediate filament proteins that form the nuclear lamina. Point mutations in genes that encode a specific subset of lamins, the A-type lamins, cause a spectrum of diseases termed laminopathies. Recent evidence points to a role for A-type lamins in intracellular redox homeostasis. To determine whether lamin A/C depletion and prelamin A accumulation differentially induce oxidative stress, we have performed a quantitative microscopy-based analysis of reactive oxygen species (ROS) levels and mitochondrial membrane potential (Δψm) in human fibroblasts subjected to sustained siRNA-mediated knockdown of LMNA and ZMPSTE24, respectively. We measured a highly significant increase in basal ROS levels and an even more prominent rise of induced ROS levels in lamin A/C depleted cells, eventually resulting in Δψm hyperpolarization and apoptosis. Depletion of ZMPSTE24 on the other hand, triggered a senescence pathway that was associated with moderately increased ROS levels and a transient Δψm depolarization. Both knockdowns were accompanied by an upregulation of several ROS detoxifying enzymes. Taken together, our data suggest that both persistent prelamin A accumulation and lamin A/C depletion elevate ROS levels, but to a different extent and with different effects on cell fate. This may contribute to the variety of disease phenotypes witnessed in laminopathies.
Collapse
Key Words
- CM-H2DCFDA, 5-(and-6)-chloromethyl-2′,7′-dichlorodihydrofluorescein diacetate
- HGPS, Hutchinson-Gilford progeria syndrome
- LA, lamin A
- LMNAkd, LMNA knockdown
- MEF, mouse embryonic fibroblasts
- NHDF, normal human dermal fibroblasts
- NT, non-targeting
- OCR, oxygen consumtion rate
- PDL, population doubling level
- PLA, prelamin A
- RD, restrictive dermopathy
- ROS, reactive oxygen species
- TBHP, tert-butyl hydrogen peroxide
- TMRM, tetramethyl rhodamine methyl ester
- ZMPSTE24
- ZMPSTE24kd, ZMPSTE24 knockdown
- apoptosis
- hMSCs, human mesenchymal stem cells
- high-content microscopy
- lamin A/C
- laminopathies
- mitochondria
- mitochondrial dysfunction
- oxidative stress
- prelamin A
- senescence
- Δψm, mitochondrial membrane potential
Collapse
Affiliation(s)
- Tom Sieprath
- a Laboratory of Cell Biology and Histology; Department of Veterinary Sciences ; University of Antwerp ; Antwerp , Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|