1
|
Saharan K, Baral S, Gandhi S, Singh AK, Ghosh S, Das R, Nagaraj VA, Vasudevan D. Structure-function studies of a nucleoplasmin isoform from Plasmodium falciparum. J Biol Chem 2025; 301:108379. [PMID: 40049416 PMCID: PMC11993163 DOI: 10.1016/j.jbc.2025.108379] [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: 07/28/2024] [Revised: 02/14/2025] [Accepted: 02/27/2025] [Indexed: 04/01/2025] Open
Abstract
An organized regulation of gene expression and DNA replication is vital for the progression of the complex life cycle of Plasmodium falciparum (Pf), involving multiple hosts and various stages. These attributes rely on the dynamic architecture of chromatin governed by several factors, including histone chaperones. Nucleoplasmin class of histone chaperones perform histone chaperoning function and participate in various developmental processes in eukaryotes. Here, our crystal structure confirmed that Pf indeed possesses a nucleoplasmin isoform (PfNPM), and the N-terminal core domain (NTD) adopts the characteristic pentameric doughnut conformation. Furthermore, PfNPM exists as a pentamer in solution, and the N-terminal core domain exhibits thermal and chemical stability. PfNPM interacts individually with assembled H2A/H2B and H3/H4 with an equimolar stoichiometry, wherein the acidic tracts of PfNPM were found to be necessary for these interactions. Further, H3/H4 displays a higher binding affinity for PfNPM than H2A/H2B, potentially due to stronger electrostatic interactions. The interaction studies also suggested that H2A/H2B and H3/H4 might share the same binding site on the PfNPM distal face, wherein H3/H4 could substitute H2A/H2B due to a higher binding affinity. Intriguingly, PfNPM neither demonstrated direct interaction with the nucleosome core particles nor displayed nucleosome assembly function, suggesting it may not be directly associated with histone deposition on the parasite genomic DNA. Furthermore, our immunofluorescence results suggested that PfNPM predominantly localizes in the nucleus and exhibits expression only in the early blood stages, such as ring and trophozoite. Altogether, we provide the first report on the structural and functional characterization of PfNPM.
Collapse
Affiliation(s)
- Ketul Saharan
- Structural Biology Laboratory, BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar, India; Regional Centre for Biotechnology, Faridabad, India
| | - Somanath Baral
- Structural Biology Laboratory, BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar, India
| | - Surajit Gandhi
- Structural Biology Laboratory, BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar, India; Regional Centre for Biotechnology, Faridabad, India
| | - Ajit Kumar Singh
- Structural Biology Laboratory, BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar, India
| | - Sourav Ghosh
- Malaria Parasite Biology Laboratory, BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar, India
| | - Rahul Das
- Malaria Parasite Biology Laboratory, BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar, India; Regional Centre for Biotechnology, Faridabad, India
| | - Viswanathan Arun Nagaraj
- Malaria Parasite Biology Laboratory, BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar, India
| | - Dileep Vasudevan
- Structural Biology Laboratory, BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar, India; Structural Biology Laboratory, BRIC-Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, India.
| |
Collapse
|
2
|
Falini B, Sorcini D, Perriello VM, Sportoletti P. Functions of the native NPM1 protein and its leukemic mutant. Leukemia 2025; 39:276-290. [PMID: 39690184 DOI: 10.1038/s41375-024-02476-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024]
Abstract
The nucleophosmin (NPM1) gene encodes for the most abundant nucleolar protein. Thanks to its property to act as histone chaperone and to shuttle between the nucleus and cytoplasm, the NPM1 protein is involved in multiple cellular function that are here extensively reviewed and include the formation of the nucleolus through liquid-liquid phase separation, regulation of ribosome biogenesis and transport, control of DNA repair and centrosome duplication as well as response to nucleolar stress. NPM1 is mutated in about 30-35% of adult acute myeloid leukemia (AML). Due to its unique biological and clinical features, NPM1-mutated AML is regarded as a distinct leukemia entity in the WHO 5th edition and ICC classifications of myeloid malignancies. The NPM1 mutant undergoes changes at the C-terminus of the protein that leads to its delocalization in the cytoplasm of the leukemic cells. Here, we focus also on its biological functions discussing the murine models of NPM1 mutations and the various mechanisms that occur at cytoplasmic and nuclear levels to promote and maintain NPM1-mutated AML.
Collapse
Affiliation(s)
- Brunangelo Falini
- Institute of Hematology and Center for Hemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy.
| | - Daniele Sorcini
- Institute of Hematology and Center for Hemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Vincenzo Maria Perriello
- Institute of Hematology and Center for Hemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Paolo Sportoletti
- Institute of Hematology and Center for Hemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
3
|
Biswal N, Harish R, Roshan M, Samudrala S, Jiao X, Pestell RG, Ashton AW. Role of GPCR Signaling in Anthracycline-Induced Cardiotoxicity. Cells 2025; 14:169. [PMID: 39936961 PMCID: PMC11817789 DOI: 10.3390/cells14030169] [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: 10/18/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 02/13/2025] Open
Abstract
Anthracyclines are a class of chemotherapeutics commonly used to treat a range of cancers. Despite success in improving cancer survival rates, anthracyclines have dose-limiting cardiotoxicity that prevents more widespread clinical utility. Currently, the therapeutic options for these patients are limited to the iron-chelating agent dexrazoxane, the only FDA-approved drug for anthracycline cardiotoxicity. However, the clinical use of dexrazoxane has failed to replicate expectations from preclinical studies. A limited list of GPCRs have been identified as pathogenic in anthracycline-induced cardiotoxicity, including receptors (frizzled, adrenoreceptors, angiotensin II receptors) previously implicated in cardiac remodeling in other pathologies. The RNA sequencing of iPSC-derived cardiac myocytes from patients has increased our understanding of the pathogenic mechanisms driving cardiotoxicity. These data identified changes in the expression of novel GPCRs, heterotrimeric G proteins, and the regulatory pathways that govern downstream signaling. This review will capitalize on insights from these experiments to explain aspects of disease pathogenesis and cardiac remodeling. These data provide a cornucopia of possible unexplored potential pathways by which we can reduce the cardiotoxic side effects, without compromising the anti-cancer effects, of doxorubicin and provide new therapeutic options to improve the recovery and quality of life for patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Nimish Biswal
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
| | - Ritika Harish
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
| | - Minahil Roshan
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
| | - Sathvik Samudrala
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
| | - Xuanmao Jiao
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
| | - Richard G. Pestell
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Anthony W. Ashton
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
- Division of Perinatal Research, Kolling Institute of Medical Research, University of Sydney, St Leonards, NSW 2065, Australia
- Division of Cardiovascular Medicine, Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| |
Collapse
|
4
|
Fu W, Zhang M, Meng Y, Wang J, Sun L. Increased NPM1 inhibit ferroptosis and aggravate renal fibrosis via Nrf2 pathway in chronic kidney disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167551. [PMID: 39437857 DOI: 10.1016/j.bbadis.2024.167551] [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: 05/04/2024] [Revised: 09/29/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Recent findings underscore the significance of ferroptosis, an innovative iron-dependent mode of cell death, in the etiology and progression of chronic kidney disease (CKD). Nucleophosmin 1 (NPM1), a nucleolar protein, contributes to fibrogenesis and modulates cellular functions and mortality. Initial investigations utilized bioinformatics techniques to pinpoint genes with altered expression in CKD and to forecast the potential links between NPM1, ferroptosis, and renal fibrosis. Increased NPM1 expression was verified in the renal tissues of CKD patients. Experimental models of renal fibrosis in both animals and cells were then used for further study. The suppression of NPM1 led to an augmentation in iron metabolism and lipid peroxidation processes integral to ferroptosis, contributing to the mitigation of renal fibrosis. In contrast, an elevation in NPM1 expression had the opposite effect. This modulation may be interconnected with the nuclear factor erythroid 2-related factor 2 pathway. Moreover, the application of the ferroptosis inhibitor, Fer-1, not only obstructed ferroptosis but also diminished NPM1 expression, which, in turn, contributed to the alleviation of renal fibrosis. Thus, our findings suggest that in CKD the NPM1 level increased and led to decreased ferroptosis and aggravated renal fibrosis via an Nrf2 pathway. Ferroptosis inhibitor can alleviate renal fibrosis.
Collapse
Affiliation(s)
- Wenjing Fu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Mingyu Zhang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Yilin Meng
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Jingyu Wang
- Renal Division, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Li Sun
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.
| |
Collapse
|
5
|
Han X, Jiang Z, Hou Y, Zhou X, Hu B. Myocardial ischemia-reperfusion injury upregulates nucleostemin expression via HIF-1α and c-Jun pathways and alleviates apoptosis by promoting autophagy. Cell Death Discov 2024; 10:461. [PMID: 39477962 PMCID: PMC11525682 DOI: 10.1038/s41420-024-02221-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 10/06/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury, often arising from interventional therapy for acute myocardial infarction, leads to irreversible myocardial cell death. While previous studies indicate that nucleostemin (NS) is induced by myocardial I/R injury and mitigates myocardial cell apoptosis, the underlying mechanisms are poorly understood. Here, our study reveals that NS upregulation is critical for preventing cardiomyocyte death following myocardial I/R injury. Elevated NS protein levels were observed in myocardial I/R injury mouse and rat models, as well as Hypoxia/reoxygenation (H/R) cardiac cell lines (H9C2 cells). We identified binding sites for c-Jun and HIF-1α in the NS promoter region. Inhibition of JNK and HIF-1α led to a significant decrease in NS transcription and protein expression. Furthermore, inhibition of autophagy and NS expression promoted myocardial cell apoptosis in H/R. Notably, the cell model showed reduced LC3I transformation to LC3II, downregulated Beclin1, upregulated p62, and altered expression of autophagy-related proteins upon NS interference in H/R cells. These findings suggest that NS expression, driven by c-Jun and HIF-1α pathways, facilitates autophagy, providing protection against both myocardial I/R injury and H/R-induced cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Xiao Han
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University & Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Zhicheng Jiang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University & Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Yufeng Hou
- Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Xiaorong Zhou
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University & Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
- Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
| | - Baoying Hu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University & Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
- Department of Immunology, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
| |
Collapse
|
6
|
Kuang Z, Ge Y, Cao L, Wang X, Liu K, Wang J, Zhu X, Wu M, Li J. Precision Treatment of Anthracycline-Induced Cardiotoxicity: An Updated Review. Curr Treat Options Oncol 2024; 25:1038-1054. [PMID: 39066853 PMCID: PMC11329674 DOI: 10.1007/s11864-024-01238-9] [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] [Accepted: 06/19/2024] [Indexed: 07/30/2024]
Abstract
OPINION STATEMENT Anthracycline (ANT)-induced cardiotoxicity (AIC) is a particularly prominent form of cancer therapy-related cardiovascular toxicity leading to the limitations of ANTs in clinical practice. Even though AIC has drawn particular attention, the best way to treat it is remaining unclear. Updates to AIC therapy have been made possible by recent developments in research on the underlying processes of AIC. We review the current molecular pathways leading to AIC: 1) oxidative stress (OS) including enzymatic-induced and other mechanisms; 2) topoisomerase; 3) inflammatory response; 4) cardiac progenitor cell damage; 5) epigenetic changes; 6) renin-angiotensin-aldosterone system (RAAS) dysregulation. And we systematically discuss current prevention and treatment strategies and novel pathogenesis-based therapies for AIC: 1) dose reduction and change; 2) altering drug delivery methods; 3) antioxidants, dexrezosen, statina, RAAS inhibitors, and hypoglycemic drugs; 4) miRNA, natural phytochemicals, mesenchymal stem cells, and cardiac progenitor cells. We also offer a fresh perspective on the management of AIC by outlining the current dilemmas and challenges associated with its prevention and treatment.
Collapse
Affiliation(s)
- Ziyu Kuang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Yuansha Ge
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Luchang Cao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Xinmiao Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Kexin Liu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Jiaxi Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Xiaojuan Zhu
- The 3rd affiliated hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| | - Min Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China.
| | - Jie Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China.
| |
Collapse
|
7
|
Taha MS, Ahmadian MR. Nucleophosmin: A Nucleolar Phosphoprotein Orchestrating Cellular Stress Responses. Cells 2024; 13:1266. [PMID: 39120297 PMCID: PMC11312075 DOI: 10.3390/cells13151266] [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: 07/04/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Nucleophosmin (NPM1) is a key nucleolar protein released from the nucleolus in response to stress stimuli. NPM1 functions as a stress regulator with nucleic acid and protein chaperone activities, rapidly shuttling between the nucleus and cytoplasm. NPM1 is ubiquitously expressed in tissues and can be found in the nucleolus, nucleoplasm, cytoplasm, and extracellular environment. It plays a central role in various biological processes such as ribosome biogenesis, cell cycle regulation, cell proliferation, DNA damage repair, and apoptosis. In addition, it is highly expressed in cancer cells and solid tumors, and its mutation is a major cause of acute myeloid leukemia (AML). This review focuses on NPM1's structural features, functional diversity, subcellular distribution, and role in stress modulation.
Collapse
Affiliation(s)
- Mohamed S. Taha
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Research on Children with Special Needs Department, Institute of Medical Research and Clinical Studies, National Research Centre, Cairo 12622, Egypt
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
8
|
Bietar K, Chu S, Mandl G, Zhang E, Chabaytah N, Sabelli R, Capobianco JA, Stochaj U. Silica-coated LiYF 4:Yb 3+, Tm 3+ upconverting nanoparticles are non-toxic and activate minor stress responses in mammalian cells. RSC Adv 2024; 14:8695-8708. [PMID: 38495986 PMCID: PMC10938293 DOI: 10.1039/d3ra08869c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
Lanthanide-doped upconverting nanoparticles (UCNPs) are ideal candidates for use in biomedicine. The interaction of nanomaterials with biological systems determines whether they are suitable for use in living cells. In-depth knowledge of the nano-bio interactions is therefore a pre-requisite for the development of biomedical applications. The current study evaluates fundamental aspects of the NP-cell interface for square bipyramidal UCNPs containing a LiYF4:Yb3+, Tm3+ core and two different silica surface coatings. Given their importance for mammalian physiology, fibroblast and renal proximal tubule epithelial cells were selected as cellular model systems. We have assessed the toxicity of the UCNPs and measured their impact on the homeostasis of living non-malignant cells. Rigorous analyses were conducted to identify possible toxic and sub-lethal effects of the UCNPs. To this end, we examined biomarkers that reveal if UCNPs induce cell killing or stress. Quantitative measurements demonstrate that short-term exposure to the UCNPs had no profound effects on cell viability, cell size or morphology. Indicators of oxidative, endoplasmic reticulum, or nucleolar stress, and the production of molecular chaperones varied with the surface modification of the UCNPs and the cell type analyzed. These differences emphasize the importance of evaluating cells of diverse origin that are relevant to the intended use of the nanomaterials. Taken together, we established that short-term, our square bipyramidal UCNPs are not toxic to non-malignant fibroblast and proximal renal epithelial cells. Compared with established inducers of cellular stress, these UCNPs have minor effects on cellular homeostasis. Our results build the foundation to explore square bipyramidal UCNPs for future in vivo applications.
Collapse
Affiliation(s)
- Kais Bietar
- Department of Physiology, McGill University Canada
| | - Siwei Chu
- Department of Physiology, McGill University Canada
| | - Gabrielle Mandl
- Department of Chemistry and Biochemistry, Centre for Nanoscience Research, Concordia University Canada
| | - Emma Zhang
- Department of Physiology, McGill University Canada
| | | | | | - John A Capobianco
- Department of Chemistry and Biochemistry, Centre for Nanoscience Research, Concordia University Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University Canada
- Quantitative Life Sciences Program, McGill University Montreal Canada
| |
Collapse
|
9
|
Sengar AS, Kumar M, Rai C, Chakraborti S, Kumar D, Kumar P, Mukherjee S, Mondal K, Stewart A, Maity B. RGS6 drives cardiomyocyte death following nucleolar stress by suppressing Nucleolin/miRNA-21. J Transl Med 2024; 22:204. [PMID: 38409136 PMCID: PMC10895901 DOI: 10.1186/s12967-024-04985-3] [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: 11/26/2023] [Accepted: 02/12/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Prior evidence demonstrated that Regulator of G protein Signaling 6 (RGS6) translocates to the nucleolus in response to cytotoxic stress though the functional significance of this phenomenon remains unknown. METHODS Utilizing in vivo gene manipulations in mice, primary murine cardiac cells, human cell lines and human patient samples we dissect the participation of a RGS6-nucleolin complex in chemotherapy-dependent cardiotoxicity. RESULTS Here we demonstrate that RGS6 binds to a key nucleolar protein, Nucleolin, and controls its expression and activity in cardiomyocytes. In the human myocyte AC-16 cell line, induced pluripotent stem cell derived cardiomyocytes, primary murine cardiomyocytes, and the intact murine myocardium tuning RGS6 levels via overexpression or knockdown resulted in diametrically opposed impacts on Nucleolin mRNA, protein, and phosphorylation.RGS6 depletion provided marked protection against nucleolar stress-mediated cell death in vitro, and, conversely, RGS6 overexpression suppressed ribosomal RNA production, a key output of the nucleolus, and triggered death of myocytes. Importantly, overexpression of either Nucleolin or Nucleolin effector miRNA-21 counteracted the pro-apoptotic effects of RGS6. In both human and murine heart tissue, exposure to the genotoxic stressor doxorubicin was associated with an increase in the ratio of RGS6/Nucleolin. Preventing RGS6 induction via introduction of RGS6-directed shRNA via intracardiac injection proved cardioprotective in mice and was accompanied by restored Nucleolin/miRNA-21 expression, decreased nucleolar stress, and decreased expression of pro-apoptotic, hypertrophy, and oxidative stress markers in heart. CONCLUSION Together, these data implicate RGS6 as a driver of nucleolar stress-dependent cell death in cardiomyocytes via its ability to modulate Nucleolin. This work represents the first demonstration of a functional role for an RGS protein in the nucleolus and identifies the RGS6/Nucleolin interaction as a possible new therapeutic target in the prevention of cardiotoxicity.
Collapse
Affiliation(s)
- Abhishek Singh Sengar
- Centre of Biomedical Research (CBMR), SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Manish Kumar
- Centre of Biomedical Research (CBMR), SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Chetna Rai
- Centre of Biomedical Research (CBMR), SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Sreemoyee Chakraborti
- Centre of Biomedical Research (CBMR), SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
- Forensic Science Laboratory, Department of Home and Hill Affairs, Kolkata, West Bengal, 700037, India
| | - Dinesh Kumar
- Centre of Biomedical Research (CBMR), SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Pranesh Kumar
- Institute of Pharmaceutical Science, University of Lucknow, Lucknow, Uttar Pradesh, 226007, India
| | - Sukhes Mukherjee
- Biochemistry, AIIMS Bhopal, Saket Nagar, Bhopal, Madhya Pradesh, 462026, India
| | - Kausik Mondal
- Zoology, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Adele Stewart
- Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Biswanath Maity
- Centre of Biomedical Research (CBMR), SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India.
| |
Collapse
|
10
|
Çakırca G, Öztürk MT, Telkoparan-Akillilar P, Güllülü Ö, Çetinkaya A, Tazebay UH. Proteomics analysis identifies the ribosome associated coiled-coil domain-containing protein-124 as a novel interaction partner of nucleophosmin-1. Biol Cell 2024; 116:e202300049. [PMID: 38029384 DOI: 10.1111/boc.202300049] [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: 05/19/2023] [Revised: 10/18/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND INFORMATION Coiled-coil domain-containing protein-124 (Ccdc124) is a conserved eukaryotic ribosome-associated RNA-binding protein which is involved in resuming ribosome activity after stress-related translational shutdown. Ccdc124 protein is also detected at cellular localizations devoid of ribosomes, such as the centrosome, or the cytokinetic midbody, but its translation-independent cellular function is currently unknown. RESULTS By using an unbiased LC-MS/MS-based proteomics approach in human embryonic kidney (HEK293) cells, we identified novel Ccdc124 partners and mapped the cellular organization of interacting proteins, a subset of which are known to be involved in nucleoli biogenesis and function. We then identified a novel interaction between the cancer-associated multifunctional nucleolar marker nucleophosmin (Npm1) and Ccdc124, and we characterized this interaction both in HEK293 (human embryonic kidney) and U2OS (osteosarcoma) cells. As expected, in both types of cells, Npm1 and Ccdc124 proteins colocalized within the nucleolus when assayed by immunocytochemical methods, or by monitoring the localization of green fluorescent protein-tagged Ccdc124. CONCLUSIONS The nucleolar localization of Ccdc124 was impaired when Npm1 translocates from the nucleolus to the nucleoplasm in response to treatment with the DNA-intercalator and Topo2 inhibitor chemotherapeutic drug doxorubicin. Npm1 is critically involved in maintaining genomic stability by mediating various DNA-repair pathways, and over-expression of Npm1 or specific NPM1 mutations have been previously associated with proliferative diseases, such as acute myelogenous leukemia, anaplastic large-cell lymphoma, and solid cancers originating from different tissues. SIGNIFICANCE Identification of Ccdc124 as a novel interaction partner of Nmp1 within the frame of molecular mechanisms involving nucleolar stress-sensing and DNA-damage response is expected to provide novel insights into the biology of cancers associated with aberrations in NPM1.
Collapse
Affiliation(s)
- Gamze Çakırca
- Gebze Technical University, Department of Molecular Biology and Genetics, Gebze, Kocaeli, Turkey
- Gebze Technical University, Central Research Laboratory (GTU-MAR), Gebze, Kocaeli, Turkey
| | - Merve Tuzlakoğlu Öztürk
- Gebze Technical University, Department of Molecular Biology and Genetics, Gebze, Kocaeli, Turkey
- Gebze Technical University, Central Research Laboratory (GTU-MAR), Gebze, Kocaeli, Turkey
| | | | - Ömer Güllülü
- Gebze Technical University, Department of Molecular Biology and Genetics, Gebze, Kocaeli, Turkey
| | - Agit Çetinkaya
- Gebze Technical University, Department of Molecular Biology and Genetics, Gebze, Kocaeli, Turkey
- Gebze Technical University, Central Research Laboratory (GTU-MAR), Gebze, Kocaeli, Turkey
| | - Uygar Halis Tazebay
- Gebze Technical University, Department of Molecular Biology and Genetics, Gebze, Kocaeli, Turkey
- Gebze Technical University, Central Research Laboratory (GTU-MAR), Gebze, Kocaeli, Turkey
| |
Collapse
|
11
|
Liu Y, Zhang L, Chen X, Sun C, Zhang Y, Li Y, Li C. Functional characterization of porcine nucleophosmin (NPM1) gene in promoting the replication of Japanese encephalitis virus and induction of inflammatory cytokines. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 148:104902. [PMID: 37536401 DOI: 10.1016/j.dci.2023.104902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Nucleophosmin (NPM1) is a multifunctional nucleolar protein that plays a role in cell cycle control, tumorigenesis, induction of the inflammatory cytokine, virus replication, as well as the cellular responses to a variety of stress stimuli. However, its physiological functions in pigs have not been well understood. Here, we cloned the porcine NPM1 (porNPM1) gene and analyzed the functions of the porNPM1 protein in pigs. The full-length porNPM1 gene encoded a 294-amino acid protein with 94.5%-99.3% sequence identity to its orthologues in mammals and was extensively expressed in various pig tissues at the mRNA level. The porNPM1 primarily localizes in the nucleus of ST cells, while it translocates from the nucleus to nucleoplasm upon UV irradiation or H2O2 treatment. Notably, JEV infection blocked the translocation of porNPM1 from the nucleolus to the nucleoplasm. Furthermore, porNPM1 interacted with the JEV C protein and facilitated JEV replication in ST cells. The overexpression and knockdown of porNPM1 respectively enhanced or impaired JEV replication, suggesting the important role of porNPM1 in JEV replication. Additionally, the purified ectodomain of porNPM1 induced the production of inflammatory cytokines (TNF-α, IL-6, and IL-8). Together, these data demonstrated that porNPM1 is involved in cellular stress stimuli, JEV replication, and induction of inflammatory cytokines.
Collapse
Affiliation(s)
- Ying Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Linjie Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Xuan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Chuwen Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Yanbing Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China
| | - Yanhua Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Comparative Medicine Research Institute, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China.
| | - Chenxi Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Comparative Medicine Research Institute, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
12
|
Wang F, Cheng H, Zhang Q, Guo J. Genetic mutations in ribosomal biogenesis gene TCOF1 identified in human neural tube defects. Mol Genet Genomic Med 2023; 11:e2150. [PMID: 36808708 PMCID: PMC10178795 DOI: 10.1002/mgg3.2150] [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: 05/24/2022] [Revised: 12/19/2022] [Accepted: 01/25/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Rare mutations in multiple genes have been associated with human neural tube defects (NTDs), but their causative roles in NTDs disease are poorly understood. Insufficiency of the ribosomal biogenesis gene treacle ribosome biogenesis factor 1(Tcof1) results in cranial NTDs and craniofacial malformations in mice. Here, we aimed to identify genetic association of TCOF1 with human NTDs. METHODS High-throughput sequencing targeted on TCOF1 was performed on samples from 355 human cases affected by NTDs and 225 controls from a Han Chinese population. RESULTS Four novel missense variants were found in the NTD cohort. Cell-based assays indicated that the p.(A491G) variant carried by an individual, who shows anencephaly and single-nostril abnormality, attenuates production of total proteins, suggesting a loss-of-function mutation in ribosomal biogenesis. Importantly, this variant promotes nucleolar disruption and stabilizes p53 protein, highlighting an unbalancing effect on cell apoptosis. CONCLUSIONS This study explored the functional impact of a missense variant in TCOF1, implicating a set of novel causative biological factors involved in the pathogenicity of human NTDs, particularly whom combined with craniofacial abnormality.
Collapse
Affiliation(s)
- Fang Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Haiqin Cheng
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China.,Department of Biochemistry and Molecular Biology, Ministry of Education Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, China
| | - Qin Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Jin Guo
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
13
|
Sahu S, Gordon J, Gu C, Sobhany M, Fiedler D, Stanley RE, Shears SB. Nucleolar Architecture Is Modulated by a Small Molecule, the Inositol Pyrophosphate 5-InsP 7. Biomolecules 2023; 13:biom13010153. [PMID: 36671538 PMCID: PMC9855682 DOI: 10.3390/biom13010153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/28/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
Inositol pyrophosphates (PP-InsPs); are a functionally diverse family of eukaryotic molecules that deploy a highly-specialized array of phosphate groups as a combinatorial cell-signaling code. One reductive strategy to derive a molecular-level understanding of the many actions of PP-InsPs is to individually characterize the proteins that bind them. Here, we describe an alternate approach that seeks a single, collective rationalization for PP-InsP binding to an entire group of proteins, i.e., the multiple nucleolar proteins previously reported to bind 5-InsP7 (5-diphospho-inositol-1,2,3,4,6-pentakisphosphate). Quantitative confocal imaging of the outer nucleolar granular region revealed its expansion when cellular 5-InsP7 levels were elevated by either (a) reducing the 5-InsP7 metabolism by a CRISPR-based knockout (KO) of either NUDT3 or PPIP5Ks; or (b), the heterologous expression of wild-type inositol hexakisphosphate kinase, i.e., IP6K2; separate expression of a kinase-dead IP6K2 mutant did not affect granular volume. Conversely, the nucleolar granular region in PPIP5K KO cells shrank back to the wild-type volume upon attenuating 5-InsP7 synthesis using either a pan-IP6K inhibitor or the siRNA-induced knockdown of IP6K1+IP6K2. Significantly, the inner fibrillar volume of the nucleolus was unaffected by 5-InsP7. We posit that 5-InsP7 acts as an 'electrostatic glue' that binds together positively charged surfaces on separate proteins, overcoming mutual protein-protein electrostatic repulsion the latter phenomenon is a known requirement for the assembly of a non-membranous biomolecular condensate.
Collapse
Affiliation(s)
- Soumyadip Sahu
- Inositol Signaling Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Jacob Gordon
- Nucleolar Integrity Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Keith Peters Building, Hills Rd, Cambridge CB2 0XY, UK
- Department of Haematology, University of Cambridge School of Clinical Medicine, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
- Wellcome Trust-Medical Research Council Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Chunfang Gu
- Inositol Signaling Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Mack Sobhany
- Nucleolar Integrity Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Dorothea Fiedler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Robin E. Stanley
- Nucleolar Integrity Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Stephen B. Shears
- Inositol Signaling Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
- Correspondence: ; Tel.: +1-984-287-3483
| |
Collapse
|
14
|
Boussada M, Hammami I, Ben Ali R, Ammar AB, Alves M, Oliveira PF, Akacha AB, Abdelkarim IL, Zekri S, El May MV. In vivo exposure to a new 2‐cyano‐2‐
p
‐nitrophenyl‐
N
‐benzylthioamide decreases doxorubicin‐triggered structural damages in the mature testis. Andrologia 2022; 54:e14634. [DOI: 10.1111/and.14634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 06/16/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Marwa Boussada
- Research Unit 17/ES/13, Laboratory of Histology and Embryology, Faculty of Medicine of Tunis University of Tunis El Manar Tunis Tunisia
| | - Imen Hammami
- Research Unit 17/ES/13, Laboratory of Histology and Embryology, Faculty of Medicine of Tunis University of Tunis El Manar Tunis Tunisia
| | - Ridha Ben Ali
- Research Unit 17/ES/13, Laboratory of Histology and Embryology, Faculty of Medicine of Tunis University of Tunis El Manar Tunis Tunisia
- Experimental Medicine Unit, Faculty of Medicine of Tunis University of Tunis El Manar Tunis Tunisia
| | - Awatef Ben Ammar
- Laboratory of Electronic Microscopy, Faculty of Medicine of Tunis University of Tunis El Manar Tunis Tunisia
| | - Marco Alves
- Department of Microscopy, Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS) University of Porto Porto Portugal
| | - Pedro Fontes Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS) University of Porto Porto Portugal
- i3S, Instituto de Investigação e Inovação da Universidade do Porto Porto Portugal
- Department of Genetics, Faculty of Medicine University of Porto Porto Portugal
| | - Azaiez Ben Akacha
- Laboratory of Organic Synthesis and Heterocyclic Chemistry Department, School of Sciences of Tunis University of Tunis El Manar Tunis Tunisia
| | - Ines Limam Abdelkarim
- Laboratory oh Hematology, Faculty of Medicine of Tunis University of Tunis El Manar Tunis Tunisia
| | - Sami Zekri
- Laboratory of Electronic Microscopy, Faculty of Medicine of Tunis University of Tunis El Manar Tunis Tunisia
| | - Michèle Véronique El May
- Research Unit 17/ES/13, Laboratory of Histology and Embryology, Faculty of Medicine of Tunis University of Tunis El Manar Tunis Tunisia
| |
Collapse
|
15
|
Yan D, Hua L. Nucleolar stress: Friend or foe in cardiac function? Front Cardiovasc Med 2022; 9:1045455. [PMID: 36386352 PMCID: PMC9659567 DOI: 10.3389/fcvm.2022.1045455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 03/14/2024] Open
Abstract
Studies in the past decades have uncovered an emerging role of the nucleolus in stress response and human disease progression. The disruption of ribosome biogenesis in the nucleolus causes aberrant nucleolar architecture and function, termed nucleolar stress, to initiate stress-responsive pathways via nucleolar release sequestration of various proteins. While data obtained from both clinical and basic investigations have faithfully demonstrated an involvement of nucleolar stress in the pathogenesis of cardiomyopathy, much remains unclear regarding its precise role in the progression of cardiac diseases. On the one hand, the initiation of nucleolar stress following acute myocardial damage leads to the upregulation of various cardioprotective nucleolar proteins, including nucleostemin (NS), nucleophosmin (NPM) and nucleolin (NCL). As a result, nucleolar stress plays an important role in facilitating the survival and repair of cardiomyocytes. On the other hand, abnormalities in nucleolar architecture and function are correlated with the deterioration of cardiac diseases. Notably, the cardiomyocytes of advanced ischemic and dilated cardiomyopathy display impaired silver-stained nucleolar organiser regions (AgNORs) and enlarged nucleoli, resembling the characteristics of tissue aging. Collectively, nucleolar abnormalities are critically involved in the development of cardiac diseases.
Collapse
Affiliation(s)
- Daliang Yan
- Department of Cardiovascular Surgery, Taizhou People’s Hospital, Taizhou, China
| | - Lu Hua
- Department of Oncology, Taizhou People’s Hospital, Taizhou, China
| |
Collapse
|
16
|
Nucleolus and Nucleolar Stress: From Cell Fate Decision to Disease Development. Cells 2022; 11:cells11193017. [PMID: 36230979 PMCID: PMC9563748 DOI: 10.3390/cells11193017] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
Besides the canonical function in ribosome biogenesis, there have been significant recent advances towards the fascinating roles of the nucleolus in stress response, cell destiny decision and disease progression. Nucleolar stress, an emerging concept describing aberrant nucleolar structure and function as a result of impaired rRNA synthesis and ribosome biogenesis under stress conditions, has been linked to a variety of signaling transductions, including but not limited to Mdm2-p53, NF-κB and HIF-1α pathways. Studies have uncovered that nucleolus is a stress sensor and signaling hub when cells encounter various stress conditions, such as nutrient deprivation, DNA damage and oxidative and thermal stress. Consequently, nucleolar stress plays a pivotal role in the determination of cell fate, such as apoptosis, senescence, autophagy and differentiation, in response to stress-induced damage. Nucleolar homeostasis has been involved in the pathogenesis of various chronic diseases, particularly tumorigenesis, neurodegenerative diseases and metabolic disorders. Mechanistic insights have revealed the indispensable role of nucleolus-initiated signaling in the progression of these diseases. Accordingly, the intervention of nucleolar stress may pave the path for developing novel therapies against these diseases. In this review, we systemically summarize recent findings linking the nucleolus to stress responses, signaling transduction and cell-fate decision, set the spotlight on the mechanisms by which nucleolar stress drives disease progression, and highlight the merit of the intervening nucleolus in disease treatment.
Collapse
|
17
|
Sazonova EN, Gusev IA, Malofey YB, Lanshakova AV, Vdovenko SV. Effects of Neonatal Administration of Non-Opiate Analogues of Leu-Enkephalin to Heart Tissue Homeostasis of Prepubertal Albino Rats Exposed to Hypoxia. Bull Exp Biol Med 2022; 173:188-192. [PMID: 35737163 DOI: 10.1007/s10517-022-05516-0] [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: 11/12/2021] [Indexed: 10/17/2022]
Abstract
Hypobaric hypoxia (pO2 65 mm Hg, duration 4 h) induced a significant increase in the number of cardiomyocytes expressing р53, beclin-1, endothelial NO synthase and accumulation and degranulation of mast cells in the epicardium in hearts of prepubertal female rats (age 45-47 days); the number of cardiomyocytes with nucleoli decreased, while the number of single-nucleolar cardiomyocytes increased after this exposure. Five-fold administration of non-opiate analogue of leu-enkephalin (NALE peptide: Phe-D-Ala-Gly-Phe-Leu-Arg; 100 μg/kg) during the neonatal period reduced the severity of the post-hypoxic changes in the heart. Neonatal administration of NALE (100 μg/kg) against the background of NO synthase blockade with L-NAME (50 mg/kg) did not abolish the cardioprotective effects of the peptide. A similar correction of posthypoxic changes in the heart was observed after neonatal administration of original peptide G (Phe-D-Ala-Gly-Phe-Leu-Gly; 100 μg/kg). Thus, NO synthase-NO system and C-terminal amino acid Arg in the molecule of non-opiate analogue of leu-enkephalin are not required for the cardioprotective effects of peptides. Non-opiate leu-enkephalin analogs, peptides NALE and G, can be considered as promising substances for increasing heart resistance to hypoxia during later age periods.
Collapse
Affiliation(s)
- E N Sazonova
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia.
- Khabarovsk Branch of the Far Eastern Research Center for Physiology and Pathology of Respiration - Research Institute for the Protection of Motherhood and Childhood, Khabarovsk, Russia.
| | - I A Gusev
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
| | - Yu B Malofey
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
| | - A V Lanshakova
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
| | - S V Vdovenko
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
| |
Collapse
|
18
|
Boteanu RM, Suica VI, Uyy E, Ivan L, Cerveanu-Hogas A, Mares RG, Simionescu M, Schiopu A, Antohe F. Short-Term Blockade of Pro-Inflammatory Alarmin S100A9 Favorably Modulates Left Ventricle Proteome and Related Signaling Pathways Involved in Post-Myocardial Infarction Recovery. Int J Mol Sci 2022; 23:ijms23095289. [PMID: 35563680 PMCID: PMC9103348 DOI: 10.3390/ijms23095289] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 02/01/2023] Open
Abstract
Prognosis after myocardial infarction (MI) varies greatly depending on the extent of damaged area and the management of biological processes during recovery. Reportedly, the inhibition of the pro-inflammatory S100A9 reduces myocardial damage after MI. We hypothesize that a S100A9 blockade induces changes of major signaling pathways implicated in post-MI healing. Mass spectrometry-based proteomics and gene analyses of infarcted mice left ventricle were performed. The S100A9 blocker (ABR-23890) was given for 3 days after coronary ligation. At 3 and 7 days post-MI, ventricle samples were analyzed versus control and Sham-operated mice. Blockade of S100A9 modulated the expressed proteins involved in five biological processes: leukocyte cell–cell adhesion, regulation of the muscle cell apoptotic process, regulation of the intrinsic apoptotic signaling pathway, sarcomere organization and cardiac muscle hypertrophy. The blocker induced regulation of 36 proteins interacting with or targeted by the cellular tumor antigen p53, prevented myocardial compensatory hypertrophy, and reduced cardiac markers of post-ischemic stress. The blockade effect was prominent at day 7 post-MI when the quantitative features of the ventricle proteome were closer to controls. Blockade of S100A9 restores key biological processes altered post-MI. These processes could be valuable new pharmacological targets for the treatment of ischemic heart. Mass spectrometry data are available via ProteomeXchange with identifier PXD033683.
Collapse
Affiliation(s)
- Raluca Maria Boteanu
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Viorel-Iulian Suica
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Elena Uyy
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Luminita Ivan
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Aurel Cerveanu-Hogas
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Razvan Gheorghita Mares
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (R.G.M.); (A.S.)
| | - Maya Simionescu
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Alexandru Schiopu
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (R.G.M.); (A.S.)
- Department of Clinical Sciences Malmö, Lund University, 21428 Malmö, Sweden
| | - Felicia Antohe
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
- Correspondence: ; Tel.: +40-213-192-737
| |
Collapse
|
19
|
Efremov YM, Suter DM, Timashev PS, Raman A. 3D nanomechanical mapping of subcellular and sub-nuclear structures of living cells by multi-harmonic AFM with long-tip microcantilevers. Sci Rep 2022; 12:529. [PMID: 35017598 PMCID: PMC8752865 DOI: 10.1038/s41598-021-04443-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
Recent developments such as multi-harmonic Atomic Force Microscopy (AFM) techniques have enabled fast, quantitative mapping of nanomechanical properties of living cells. Due to their high spatiotemporal resolution, these methods provide new insights into changes of mechanical properties of subcellular structures due to disease or drug response. Here, we propose three new improvements to significantly improve the resolution, identification, and mechanical property quantification of sub-cellular and sub-nuclear structures using multi-harmonic AFM on living cells. First, microcantilever tips are streamlined using long-carbon tips to minimize long-range hydrodynamic interactions with the cell surface, to enhance the spatial resolution of nanomechanical maps and minimize hydrodynamic artifacts. Second, simultaneous Spinning Disk Confocal Microscopy (SDC) with live-cell fluorescent markers enables the unambiguous correlation between observed heterogeneities in nanomechanical maps with subcellular structures. Third, computational approaches are then used to estimate the mechanical properties of sub-nuclear structures. Results are demonstrated on living NIH 3T3 fibroblasts and breast cancer MDA-MB-231 cells, where properties of nucleoli, a deep intracellular structure, were assessed. The integrated approach opens the door to study the mechanobiology of sub-cellular structures during disease or drug response.
Collapse
Affiliation(s)
- Yuri M Efremov
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare, Moscow, Russia
| | - Daniel M Suter
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Integrative Neuroscience, West Lafayette, IN, USA
| | - Peter S Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Arvind Raman
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA.
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
20
|
Beji S, D'Agostino M, Gambini E, Sileno S, Scopece A, Vinci MC, Milano G, Melillo G, Napolitano M, Pompilio G, Capogrossi MC, Avitabile D, Magenta A. Doxorubicin induces an alarmin-like TLR4-dependent autocrine/paracrine action of Nucleophosmin in human cardiac mesenchymal progenitor cells. BMC Biol 2021; 19:124. [PMID: 34134693 PMCID: PMC8210386 DOI: 10.1186/s12915-021-01058-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 05/28/2021] [Indexed: 12/11/2022] Open
Abstract
Background Doxorubicin (Dox) is an anti-cancer anthracycline drug that causes double-stranded DNA breaks. It is highly effective against several types of tumours; however, it also has adverse effects on regenerative populations of normal cells, such as human cardiac mesenchymal progenitor cells (hCmPCs), and its clinical use is limited by cardiotoxicity. Another known effect of Dox is nucleolar disruption, which triggers the ubiquitously expressed nucleolar phosphoprotein Nucleophosmin (NPM) to be released from the nucleolus into the cell, where it participates in the orchestration of cellular stress responses. NPM has also been observed in the extracellular space in response to different stress stimuli; however, the mechanism behind this and its functional implications are as yet largely unexplored. The aim of this study was to establish whether Dox could elicit NPM secretion in the extracellular space and to elucidate the mechanism of secretion and the effect of extracellular NPM on hCmPCs. Results We found that following the double-strand break formation in hCmPCs caused by Dox, NPM was rapidly secreted in the extracellular space by an active mechanism, in the absence of either apoptosis or necrosis. Extracellular release of NPM was similarly seen in response to ultraviolet radiation (UV). Furthermore, we observed an increase of NPM levels in the plasma of Dox-treated mice; thus, NPM release also occurred in vivo. The treatment of hCmPCs with extracellular recombinant NPM induced a decrease of cell proliferation and a response mediated through the Toll-like receptor (TLR)4. We demonstrated that NPM binds to TLR4, and via TLR4, and nuclear factor kappa B (NFkB) activation/nuclear translocation, exerts proinflammatory functions by inducing IL-6 and COX-2 gene expression. Finally, we found that in hCmPCs, NPM secretion could be driven by an autophagy-dependent unconventional mechanism that requires TLR4, since TLR4 inhibition dramatically reduced Dox-induced secretion. Conclusions We hypothesise that the extracellular release of NPM could be a general response to DNA damage since it can be elicited by either a chemical agent such as Dox or a physical genotoxic stressor such as UV radiation. Following genotoxic stress, NPM acts similarly to an alarmin in hCmPCs, being rapidly secreted and promoting cell cycle arrest and a TLR4/NFκB-dependent inflammatory response. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01058-5.
Collapse
Affiliation(s)
- Sara Beji
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Marco D'Agostino
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Elisa Gambini
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Sara Sileno
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Alessandro Scopece
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Maria Cristina Vinci
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | - Giuseppina Milano
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy
| | | | | | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Maurizio C Capogrossi
- Laboratory of Cardiovascular Science, National Institute on Aging (NIA), National Institutes of Health (NIH), 251 Bayview Blvd, Baltimore, MD, 21224, USA.,Division of Cardiology, Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Daniele Avitabile
- Idi Farmaceutici S.r.l., Via dei Castelli Romani 83/85, 00071, Pomezia (Rome), Italy.
| | - Alessandra Magenta
- National Research Council of Italy (CNR), Institute of Translational Pharmacology IFT, Via Fosso del Cavaliere 100, 00133, Rome, Italy.
| |
Collapse
|
21
|
SIRT7-dependent deacetylation of NPM promotes p53 stabilization following UV-induced genotoxic stress. Proc Natl Acad Sci U S A 2021; 118:2015339118. [PMID: 33495326 DOI: 10.1073/pnas.2015339118] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Adaptation to different forms of environmental stress is crucial for maintaining essential cellular functions and survival. The nucleolus plays a decisive role as a signaling hub for coordinating cellular responses to various extrinsic and intrinsic cues. p53 levels are normally kept low in unstressed cells, mainly due to E3 ubiquitin ligase MDM2-mediated degradation. Under stress, nucleophosmin (NPM) relocates from the nucleolus to the nucleoplasm and binds MDM2, thereby preventing degradation of p53 and allowing cell-cycle arrest and DNA repair. Here, we demonstrate that the mammalian sirtuin SIRT7 is an essential component for the regulation of p53 stability during stress responses induced by ultraviolet (UV) irradiation. The catalytic activity of SIRT7 is substantially increased upon UV irradiation through ataxia telangiectasia mutated and Rad3 related (ATR)-mediated phosphorylation, which promotes efficient deacetylation of the SIRT7 target NPM. Deacetylation is required for stress-dependent relocation of NPM into the nucleoplasm and MDM2 binding, thereby preventing ubiquitination and degradation of p53. In the absence of SIRT7, stress-dependent stabilization of p53 is abrogated, both in vitro and in vivo, impairing cellular stress responses. The study uncovers an essential SIRT7-dependent mechanism for stabilization of the tumor suppressor p53 in response to genotoxic stress.
Collapse
|
22
|
Di Carlo A, Beji S, Palmerio S, Picozza M, D’Agostino M, Petrozza V, Melchionna R, Germani A, Magenta A, De Falco E, Avitabile D. The Nucleolar Protein Nucleophosmin Is Physiologically Secreted by Endothelial Cells in Response to Stress Exerting Proangiogenic Activity Both In Vitro and In Vivo. Int J Mol Sci 2021; 22:ijms22073672. [PMID: 33916025 PMCID: PMC8037380 DOI: 10.3390/ijms22073672] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/25/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023] Open
Abstract
Nucleophosmin (NPM), a nucleolar multifunctional phosphoprotein, acts as a stress sensor in different cell types. NPM can be actively secreted by inflammatory cells, however its biology on endothelium remains unexplored. In this study, we show for the first time that NPM is secreted by human vein endothelial cells (HUVEC) in the early response to serum deprivation and that NPM acts as a pro-inflammatory and angiogenic molecule both in vitro and in vivo. Accordingly, 24 h of serum starvation condition induced NPM relocalization from the nucleus to cytoplasm. Interestingly, NPM was increasingly excreted in HUVEC-derived conditioned media in a time dependent fashion upon stress conditions up to 24 h. The secretion of NPM was unrelated to cell necrosis within 24 h. The treatment with exogenous and recombinant NPM (rNPM) enhanced migration as well as the Intercellular Adhesion Molecule 1 (ICAM-1) but not Vascular cell adhesion protein 1 (VCAM-1) expression and it did not affect cell proliferation. Notably, in vitro tube formation by Matrigel assay was significantly increased in HUVEC treated with rNPM compared to controls. This result was confirmed by the in vivo injection of Matrigel plug assay upon stimulation with rNPM, displaying significant enhanced number of functional capillaries in the plugs. The stimulation with rNPM in HUVEC was also associated to the increased expression of master genes regulating angiogenesis and migration, including Vascular Endothelial Growth Factor-A (VEGF-A), Hepatocyte Growth Factor (HGF), Stromal derived factor-1 (SDF-1), Fibroblast growth factor-2 (FGF-2), Platelet Derived Growth Factor-B (PDGF-B), and Matrix metallopeptidase 9 (MMP9). Our study demonstrates for the first time that NPM is physiologically secreted by somatic cells under stress condition and in the absence of cell necrosis. The analysis of the biological effects induced by NPM mainly related to a pro-angiogenic and inflammatory activity might suggest an important autocrine/paracrine role for NPM in the regulation of both phenomena.
Collapse
Affiliation(s)
- Anna Di Carlo
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.D.C.); (R.M.)
| | - Sara Beji
- Laboratory of Experimental Immunology, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy; (S.B.); (S.P.); (M.D.); (A.G.)
| | - Silvia Palmerio
- Laboratory of Experimental Immunology, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy; (S.B.); (S.P.); (M.D.); (A.G.)
| | - Mario Picozza
- Neuroimmunology Unit, IRCSS Fondazione Santa Lucia, 00143 Rome, Italy;
| | - Marco D’Agostino
- Laboratory of Experimental Immunology, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy; (S.B.); (S.P.); (M.D.); (A.G.)
| | - Vincenzo Petrozza
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy; (V.P.); (E.D.F.)
| | - Roberta Melchionna
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.D.C.); (R.M.)
| | - Antonia Germani
- Laboratory of Experimental Immunology, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy; (S.B.); (S.P.); (M.D.); (A.G.)
| | - Alessandra Magenta
- Institute of Translational Pharmacology (IFT), Consiglio Nazionale delle Ricerche (CNR), 00133 Rome, Italy;
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy; (V.P.); (E.D.F.)
- Mediterranea Cardiocentro, 80122 Naples, Italy
| | - Daniele Avitabile
- Department of Scientifico e Sviluppo, IDI Farmaceutici, Via dei Castelli Romani 73/75, 00071 Pomezia, Italy
- Correspondence: ; Tel.: +06-91092610
| |
Collapse
|
23
|
Huo CY, Chang ML, Cheng H, Ma TT, Fu Y, Wang Y, Wang YY, Kan YC, Li DD. Small nucleolar RNA of silkworm can translocate from the nucleolus to the cytoplasm under abiotic stress. Cell Biol Int 2021; 45:1091-1097. [PMID: 33501699 DOI: 10.1002/cbin.11555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/24/2021] [Indexed: 12/19/2022]
Abstract
Small nucleolar RNAs (snoRNAs) are thought to be exclusively nuclear and guide nucleotide modifications of ribosomal RNAs. Recently, more and more evidence has suggested that the nucleolus is a stress sensor for changes in growth status and that snoRNAs may orchestrate the response to environmental stress through molecular interactions outside of the nucleus. We previously showed that a box C/D snoRNA Bm-15 had both nuclear and cytoplasmic location in BmN4 cell line of the silkworm, Bombyx mori. To further study the functional roles of Bm-15, changes in expression level and cellular location of Bm-15 were examined in BmN4 cells subjected to serum starvation and ultraviolet (UV) ray radiation. Results indicated that total RNA level of Bm-15 was unchanged after 24 h serum starvation, but exhibited 3-fold increases in the cytoplasm, and the nuclear-to-cytosolic distribution ratio was reduced from 5:1 to 2:1. Moreover, UV radiation also causes rapid decline in nuclear Bm-15 and progressive cytoplasmic accumulation with a percentage of 22% and 57% after 6 and 24 h UV radiation. UV treatment results in a dramatic decrease in Bm-15 nuclear-to-cytosolic ratio from 7:1 to 2:1 and 2:1 to 1:20 after 6 and 24 h UV radiation, respectively. We show here for the first time that box C/D snoRNAs can translocate from the nucleus to the cytoplasm under the abiotic stress of nutritional deficiency and UV radiation. The rapid translocation of snoRNAs from nucleus to cytoplasm may slow down the maturation of rRNAs and synthesis of ribosomes to enhance the stress resistance of cells.
Collapse
Affiliation(s)
- Chun-Yue Huo
- China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 1638 Wolong Road, Nanyang, Henan, 473061, China
| | - Mei-Ling Chang
- China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 1638 Wolong Road, Nanyang, Henan, 473061, China
| | - Hao Cheng
- China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 1638 Wolong Road, Nanyang, Henan, 473061, China
| | - Tian-Tian Ma
- China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 1638 Wolong Road, Nanyang, Henan, 473061, China
| | - Yu Fu
- China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 1638 Wolong Road, Nanyang, Henan, 473061, China
| | - Yi Wang
- China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 1638 Wolong Road, Nanyang, Henan, 473061, China
| | - Yan-Yan Wang
- China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 1638 Wolong Road, Nanyang, Henan, 473061, China
| | - Yun-Chao Kan
- China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 1638 Wolong Road, Nanyang, Henan, 473061, China
- School of Life Science, Henan University, Jin Ming Avenue, Kaifeng, Henan, 475004, China
| | - Dan-Dan Li
- China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 1638 Wolong Road, Nanyang, Henan, 473061, China
| |
Collapse
|
24
|
Rao C, Liu B, Huang D, Chen R, Huang K, Li F, Dong N. Nucleophosmin contributes to vascular inflammation and endothelial dysfunction in atherosclerosis progression. J Thorac Cardiovasc Surg 2019; 161:e377-e393. [PMID: 32007256 DOI: 10.1016/j.jtcvs.2019.10.152] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/15/2019] [Accepted: 10/15/2019] [Indexed: 01/27/2023]
Abstract
OBJECTIVE It is unclear whether nucleophosmin (NPM) participates in cardiovascular disease. The present study aimed to investigate the role and underlying mechanisms of NPM in atherosclerosis. METHODS Levels and location of NPM in human carotid atherosclerotic plaques and healthy controls were detected by real-time polymerase chain reaction, immunoblots, and immunofluorescence. Atherosclerotic prone ApoE-/- mice were fed with a Western diet for 16 weeks as an in vivo model. Human primary umbilical vein endothelial cells (HUVECs) were cultured as an in vitro model. RESULTS Compared with controls, we found that NPM levels in human carotid atherosclerotic plaques were more than twice as high as in normal arteries, which mainly localized in endothelial cells. In vivo, adenovirus-containing NPM small hairpin RNA attenuated atherosclerotic lesion and promoted plaque stabilization in ApoE-/- mice fed a Western diet by reducing vascular inflammation, maintaining endothelial function, and decreasing macrophage infiltration. Furthermore, NPM knockdown decreased nuclear factor-κB (NF-κB) p65 phosphorylation. In cultured HUVECs, palmitic acid increased the protein levels of NPM and induced the expression of inflammatory cytokines and monocyte adhesion, whereas NPM knockdown attenuated this effect. In HUVECs, NPM protein physically interacted with NF-κB p65 subunit and promoted its nuclear transposition. NPM also increased the transcriptional activity of NF-κB p65 promoter and enhance its binding to target genes, including interleukin-1β, interleukin-6, intercellular adhesion molecule-1, and E-selectin. CONCLUSIONS These data provide novel evidence that NPM promotes atherosclerosis by inducing vascular inflammation and endothelial dysfunction through the NF-κB signaling pathway and suggest that NPM may be a promising target for atherosclerosis prevention and treatment.
Collapse
Affiliation(s)
- Caijun Rao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinical Center for Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baoqing Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dandan Huang
- Clinical Center for Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ru Chen
- Clinical Center for Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Clinical Center for Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Guo Q, Zhang Y, Zhang S, Jin J, Pang S, Wu X, Zhang W, Bi X, Zhang Y, Zhang Q, Jiang F. Genome-wide translational reprogramming of genes important for myocyte functions in overload-induced heart failure. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165649. [PMID: 31870714 DOI: 10.1016/j.bbadis.2019.165649] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/22/2022]
Abstract
Genome-wide changes in gene translational efficiency during the development of heart failure are poorly understood. We tested the hypothesis that aberrant changes in translational efficiency of cardiac genes are associated with the development of myocyte decompensation in response to persistent stress stimuli. We demonstrated that chronic pressure overload in mice resulted in a genome-wide reprogramming of translational efficiency, with >50% of the translatome exhibiting decreased translational efficiencies during the transition from myocardial compensation to decompensation. Importantly, these translationally repressed genes included those involved in angiogenesis and energy metabolism. Moreover, we showed that the stress-induced translational reprogramming was accompanied by persistent activation of the eukaryotic initiation factor 2α (eIF2α)-mediated stress response pathway. Counteracting the endogenous eIF2α functions by cardiac-specific overexpression of an eIF2α-S51A mutant ameliorated the development of myocyte decompensation, with concomitant improvements in translation of cardiac functional genes and increases in angiogenic responses. These data suggest that the mismatch between transcription and translation of the cardiac genes with essential functions may represent a novel molecular mechanism underlying the development of myocyte decompensation in response to chronic stress stimuli, and the eIF2α pathway may be a viable therapeutic target for recovering the optimal translation of the repressed cardiac genes.
Collapse
Affiliation(s)
- Qianqian Guo
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yongtao Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Physiology and Pathophysiology, School of Basic Medicine, Shandong University, Jinan, Shandong Province, China; Department of Cardiology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Shucui Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jiajia Jin
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shu Pang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiao Wu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wencheng Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaolei Bi
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong Province, China
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Fan Jiang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Physiology and Pathophysiology, School of Basic Medicine, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
26
|
Sazonova ЕN, Cimbalist NA, Kaplieva OV, Lebed’ko OA. The influence of non-opiate analogue of leu-enkephalin to the cardiac consequences of intrauterine hypoxia of albino rats. RUSSIAN OPEN MEDICAL JOURNAL 2019. [DOI: 10.15275/rusomj.2019.0401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Objective ― Our study aimed to evaluate the possibility of correcting cardiac consequences of intrauterine hypoxia (IUH) by injecting leu-enkephalin analog, lacking affinity for opiate receptors, in the early postnatal period. Material and Methods ― To model IUH, we placed pregnant Wistar rats in a hypobaric chamber with an oxygen partial pressure of 52 mmHg. The procedure was repeated for 4 h daily over the 15th-19th days of gestation. From the 2nd through the 6th days of their lives, the offspring were injected intraperitoneally with non-opiate leu-enkephalin analog at a dose of 100 μg/kg (NALE: Phe-D-Ala-Gly-Phe-Leu-Arg). This analog did not have affinity for opiate receptors. The 7- and 60-day old offspring of female rats subjected to IUH were investigated. The control group included the descendants of intact animals. We investigated gravimetric indicators, DNA-synthetic activity of cardiomyocytes (CMC) by tritium-labeled thymidine autoradiography method, the size of the CMC nuclei, as well as size and amount of nucleoli in the CMC nuclei. The activity of free radical oxidation was evaluated in cardiac homogenates by chemiluminescence. Results ― In 7-day old rats subjected to IUH vs. control animals, we observed decreases in body mass by 32.6%, in heart mass by 27.3%; in the proportion of 3Н-thymidine labeled CMC nuclei by 32.7% in the left ventricle and by 30.4% in the right ventricle; in the number of nucleoli in the CMC nuclei (in the left ventricle: control – 2.384±0.027, IUH – 2.282±0.027*, p<0.05; in the right ventricle: control – 2.409±0.038; IUH – 2.240±0.012*, p<0, 05). Increase in CML indices of cardiac homogenates was revealed, indicating the activation of free radical oxidation. In 7-day old rats subjected to IUH and administration of the NALE peptide from the 2nd through the 6th days of their lives, the proportion of 3H-thymidine labeled nuclei in the CMC did not differ from the control (in the left ventricle: control – 12.79±0.89%, IUH + NALE – 10.98±0.95%, p>0.05; in the right ventricle: control – 11.61±0.78%; IUH + NALE – 11.26±0.58%, p>0.05). The number of nucleoli in the CMC nuclei of the left and right ventricles in the heart of 7-day old animals in the IUH + NALE group did not differ from the control too. The CML indices of heart homogenates in the IUH + NALE group were significantly lower than those in the IUH group. In 60-day old male rats exposed to IUH, there was a decrease in heart mass by 18.5%, sizes of CMC nuclei by 7.5% and 16.1% in the left and right ventricles, respectively, and in the total nucleoli area in the CMC nuclei of the left ventricle (control – 3.953±0.085; IUH – 3.372±0.078*; p<0.05). In 60-day old male rats subjected to IUH and injections of the NALE peptide from the 2nd to the 6th days of their lives, heart mass (control – 692.73±26.81 mg; IUH + NALE – 631.0±29.79 mg; p>0.05) and the size of the CMC nuclei of the right ventricle (control – 54.25±0.84; IUH + NALE – 55.24±0.94; p>0.05) did not differ significantly from the control. The size of the nuclei, the number and size of the nucleoli in the CMC of the left ventricle, as well as the area of the nucleoli in the CMC of the right ventricle in 60-day old male rats of the IUH + NALE group significantly exceeded control group values. Conclusion ― Administration of the NALE peptide to albino rats subjected to IUH normalized DNA-synthetic activity and the number of nucleoli in the nuclei of CMC in 7-day old animals, and also reduced the severity of oxidative stress in the heart tissue. In 60-day old albino male rats exposed to IUH, injecting NALE from the 2nd to the 6th days of their lives eliminated declines in heart mass and sizes of the CMC nuclei and nucleoli, and also led to an increase in the values of the nucleus-and-nucleolus complex indices compared with the control.
Collapse
|
27
|
Hu W, Liang YX, Luo JM, Gu XW, Chen ZC, Fu T, Zhu YY, Lin S, Diao HL, Jia B, Yang ZM. Nucleolar stress regulation of endometrial receptivity in mouse models and human cell lines. Cell Death Dis 2019; 10:831. [PMID: 31685803 PMCID: PMC6828743 DOI: 10.1038/s41419-019-2071-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/20/2019] [Accepted: 10/16/2019] [Indexed: 12/17/2022]
Abstract
Embryo implantation is essential to the successful establishment of pregnancy. A previous study has demonstrated that actinomycin D (ActD) could initiate the activation of mouse delayed implantation. However, the mechanism underlying this activation remains to be elucidated. A low dose of ActD is an inducer of nucleolar stress. This study was to examine whether nucleolar stress is involved in embryo implantation. We showed that nucleolar stress occurred when delayed implantation was activated by ActD in mice. ActD treatment also stimulated the Lif-STAT3 pathway. During early pregnancy, nucleolar stress was detected in the luminal epithelial cells during the receptive phase. Blastocyst-derived lactate could induce nucleolar stress in cultured luminal epithelial cells. The inhibition of nucleophosmin1 (NPM1), which was a marker of nucleolar stress, compromised uterine receptivity and decreased the implantation rates in pregnant mice. To translate these mouse data into humans, we examined nucleolar stress in human endometrium. Our data demonstrated that ActD-induced nucleolar stress had positive effects on the embryo attachment by upregulating IL32 expression in non-receptive epithelial cells rather than receptive epithelial cells. Our data should be the first to demonstrate that nucleolar stress is present during early pregnancy and is able to induce embryo implantation in both mice and humans.
Collapse
Affiliation(s)
- Wei Hu
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China
| | - Yu-Xiang Liang
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China.,Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Animal Model of Human Disease, Shanxi Medical University, 030001, Taiyuan, China.,Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, 030001, Taiyuan, China
| | - Jia-Mei Luo
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China
| | - Xiao-Wei Gu
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China
| | - Zi-Cong Chen
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China
| | - Tao Fu
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China
| | - Yu-Yuan Zhu
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China
| | - Shuai Lin
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China
| | - Hong-Lu Diao
- Reproductive Medicine Center, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Bo Jia
- Jiangxi Provincial Institute of Occupational Medicine, 330006, Nanchang, China
| | - Zeng-Ming Yang
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China.
| |
Collapse
|
28
|
Weeks SE, Metge BJ, Samant RS. The nucleolus: a central response hub for the stressors that drive cancer progression. Cell Mol Life Sci 2019; 76:4511-4524. [PMID: 31338556 PMCID: PMC6841648 DOI: 10.1007/s00018-019-03231-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/25/2019] [Accepted: 07/15/2019] [Indexed: 01/17/2023]
Abstract
The nucleolus is a sub-nuclear body known primarily for its role in ribosome biogenesis. Increased number and/or size of nucleoli have historically been used by pathologists as a prognostic indicator of cancerous lesions. This increase in nucleolar number and/or size is classically attributed to the increased need for protein synthesis in cancer cells. However, evidences suggest that the nucleolus plays critical roles in many cellular functions in both normal cell biology and disease pathologies, including cancer. As new functions of the nucleolus are elucidated, there is mounting evidence to support the role of the nucleolus in regulating additional cellular functions, particularly response to cellular stressors, maintenance of genome stability, and DNA damage repair, as well as the regulation of gene expression and biogenesis of several ribonucleoproteins. This review highlights the central role of the nucleolus in carcinogenesis and cancer progression and discusses how cancer cells may become "addicted" to nucleolar functions.
Collapse
Affiliation(s)
- Shannon E Weeks
- Department of Pathology, University of Alabama at Birmingham, WTI 320E, 1824 6th Ave South, Birmingham, AL, 35233, USA
| | - Brandon J Metge
- Department of Pathology, University of Alabama at Birmingham, WTI 320E, 1824 6th Ave South, Birmingham, AL, 35233, USA
| | - Rajeev S Samant
- Department of Pathology, University of Alabama at Birmingham, WTI 320E, 1824 6th Ave South, Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
29
|
He JS, Soo P, Evers M, Parsons KM, Hein N, Hannan KM, Hannan RD, George AJ. High-Content Imaging Approaches to Quantitate Stress-Induced Changes in Nucleolar Morphology. Assay Drug Dev Technol 2019; 16:320-332. [PMID: 30148664 DOI: 10.1089/adt.2018.861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The nucleolus is a dynamic subnuclear compartment that has a number of different functions, but its primary role is to coordinate the production and assembly of ribosomes. For well over 100 years, pathologists have used changes in nucleolar number and size to stage diseases such as cancer. New information about the nucleolus' broader role within the cell is leading to the development of drugs which directly target its structure as therapies for disease. Traditionally, it has been difficult to develop high-throughput image analysis pipelines to measure nucleolar changes due to the broad range of morphologies observed. In this study, we describe a simple high-content image analysis algorithm using Harmony software (PerkinElmer), with a PhenoLOGIC™ machine-learning component, that can measure and classify three different nucleolar morphologies based on nucleolin and fibrillarin staining ("normal," "peri-nucleolar rings" and "dispersed"). We have utilized this algorithm to determine the changes in these classes of nucleolar morphologies over time with drugs known to alter nucleolar structure. This approach could be further adapted to include other parameters required for the identification of new therapies that directly target the nucleolus.
Collapse
Affiliation(s)
- Jin-Shu He
- 1 ANU Centre for Therapeutic Discovery, The Australian National University , Acton, Australia
| | - Priscilla Soo
- 2 ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University , Acton, Australia
| | - Maurits Evers
- 2 ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University , Acton, Australia
| | - Kate M Parsons
- 1 ANU Centre for Therapeutic Discovery, The Australian National University , Acton, Australia
| | - Nadine Hein
- 2 ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University , Acton, Australia
| | - Katherine M Hannan
- 2 ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University , Acton, Australia .,3 Department of Biochemistry and Molecular Biology, University of Melbourne , Parkville, Australia
| | - Ross D Hannan
- 2 ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University , Acton, Australia .,3 Department of Biochemistry and Molecular Biology, University of Melbourne , Parkville, Australia .,4 Sir Peter MacCallum Department of Oncology, University of Melbourne , Parkville, Australia .,5 Department of Biochemistry and Molecular Biology, University of Melbourne , Parkville, Australia .,6 Department of Biochemistry and Molecular Biology, Monash University , Clayton, Australia .,7 School of Biomedical Sciences, University of Queensland , St Lucia, Australia
| | - Amee J George
- 1 ANU Centre for Therapeutic Discovery, The Australian National University , Acton, Australia .,2 ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University , Acton, Australia .,7 School of Biomedical Sciences, University of Queensland , St Lucia, Australia .,8 Department of Clinical Pathology, University of Melbourne , Parkville, Australia
| |
Collapse
|
30
|
Abstract
Cardiac ageing manifests as a decline in function leading to heart failure. At the cellular level, ageing entails decreased replicative capacity and dysregulation of cellular processes in myocardial and nonmyocyte cells. Various extrinsic parameters, such as lifestyle and environment, integrate important signalling pathways, such as those involving inflammation and oxidative stress, with intrinsic molecular mechanisms underlying resistance versus progression to cellular senescence. Mitigation of cardiac functional decline in an ageing organism requires the activation of enhanced maintenance and reparative capacity, thereby overcoming inherent endogenous limitations to retaining a youthful phenotype. Deciphering the molecular mechanisms underlying dysregulation of cellular function and renewal reveals potential interventional targets to attenuate degenerative processes at the cellular and systemic levels to improve quality of life for our ageing population. In this Review, we discuss the roles of extrinsic and intrinsic factors in cardiac ageing. Animal models of cardiac ageing are summarized, followed by an overview of the current and possible future treatments to mitigate the deleterious effects of cardiac ageing.
Collapse
|
31
|
Bi X, Ye Q, Li D, Peng Q, Wang Z, Wu X, Zhang Y, Zhang Q, Jiang F. Inhibition of nucleolar stress response by Sirt1: A potential mechanism of acetylation-independent regulation of p53 accumulation. Aging Cell 2019; 18:e12900. [PMID: 30623565 PMCID: PMC6413664 DOI: 10.1111/acel.12900] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 11/28/2018] [Accepted: 12/08/2018] [Indexed: 02/04/2023] Open
Abstract
The mammalian Sirt1 deacetylase is generally thought to be a nuclear protein, but some pilot studies have suggested that Sirt1 may also be involved in orchestrating nucleolar functions. Here, we show that nucleolar stress response is a ubiquitous cellular reaction that can be induced by different types of stress conditions, and Sirt1 is an endogenous suppressor of nucleolar stress response. Using stable isotope labeling by amino acids in cell culture approach, we have identified a physical interaction of between Sirt1 and the nucleolar protein nucleophosmin, and this protein-protein interaction appears to be necessary for Sirt1 inhibition on nucleolar stress, whereas the deacetylase activity of Sirt1 is not strictly required. Based on the reported prerequisite role of nucleolar stress response in stress-induced p53 protein accumulation, we have also provided evidence suggesting that Sirt1-mediated inhibition on nucleolar stress response may represent a novel mechanism by which Sirt1 can modulate intracellular p53 accumulation independent of lysine deacetylation. This process may represent an alternative mechanism by which Sirt1 regulates functions of the p53 pathway.
Collapse
Affiliation(s)
- Xiaolei Bi
- School of Basic MedicineShandong UniversityJinanShandong ProvinceChina
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
- Present address:
Department of CardiologyQingdao Municipal HospitalQingdaoShandong ProvinceChina
| | - Qing Ye
- School of Basic MedicineShandong UniversityJinanShandong ProvinceChina
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Daoyuan Li
- National Glycoengineering Research CenterShandong UniversityJinanChina
| | - Qisheng Peng
- Key Laboratory of Zoonosis ResearchJilin UniversityChangchunJilin ProvinceChina
| | - Zhe Wang
- Division of Endocrinology and MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
| | - Xiao Wu
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Fan Jiang
- School of Basic MedicineShandong UniversityJinanShandong ProvinceChina
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| |
Collapse
|
32
|
Maina MB, Bailey LJ, Wagih S, Biasetti L, Pollack SJ, Quinn JP, Thorpe JR, Doherty AJ, Serpell LC. The involvement of tau in nucleolar transcription and the stress response. Acta Neuropathol Commun 2018; 6:70. [PMID: 30064522 PMCID: PMC6066928 DOI: 10.1186/s40478-018-0565-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 06/30/2018] [Indexed: 12/16/2022] Open
Abstract
Tau is known for its pathological role in neurodegenerative diseases, including Alzheimer's disease (AD) and other tauopathies. Tau is found in many subcellular compartments such as the cytosol and the nucleus. Although its normal role in microtubule binding is well established, its nuclear role is still unclear. Here, we reveal that tau localises to the nucleolus in undifferentiated and differentiated neuroblastoma cells (SHSY5Y), where it associates with TIP5, a key player in heterochromatin stability and ribosomal DNA (rDNA) transcriptional repression. Immunogold labelling on human brain sample confirms the physiological relevance of this finding by showing tau within the nucleolus colocalises with TIP5. Depletion of tau results in an increase in rDNA transcription with an associated decrease in heterochromatin and DNA methylation, suggesting that under normal conditions tau is involved in silencing of the rDNA. Cellular stress induced by glutamate causes nucleolar stress associated with the redistribution of nucleolar non-phosphorylated tau, in a similar manner to fibrillarin, and nuclear upsurge of phosphorylated tau (Thr231) which doesn't colocalise with fibrillarin or nucleolar tau. This suggests that stress may impact on different nuclear tau species. In addition to involvement in rDNA transcription, nucleolar non-phosphorylated tau also undergoes stress-induced redistribution similar to many nucleolar proteins.
Collapse
MESH Headings
- Brain/metabolism
- Brain/ultrastructure
- Cell Differentiation/physiology
- Cell Line, Tumor
- Cell Nucleolus/drug effects
- Cell Nucleolus/metabolism
- Cell Nucleolus/ultrastructure
- Chromosomal Proteins, Non-Histone/metabolism
- Chromosomal Proteins, Non-Histone/ultrastructure
- DNA, Ribosomal/genetics
- DNA, Ribosomal/metabolism
- Excitatory Amino Acid Agonists/pharmacology
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Glutamic Acid/pharmacology
- Heterochromatin/physiology
- Histones/metabolism
- Humans
- Immunoprecipitation
- Microscopy, Confocal
- Microscopy, Electron
- Neuroblastoma/pathology
- Neuroblastoma/ultrastructure
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Protein Transport/drug effects
- RNA, Messenger
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Transcription, Genetic/drug effects
- Transfection
- tau Proteins/genetics
- tau Proteins/metabolism
- tau Proteins/ultrastructure
Collapse
Affiliation(s)
- Mahmoud B Maina
- School of Life Sciences, University of Sussex, Falmer, Brighton, East Sussex, BN1 9QG, UK
- Department of Human Anatomy, College of Medical Science, Gombe State University, Gombe, Nigeria
| | - Laura J Bailey
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, BN1 9RQ, UK
| | - Sherin Wagih
- School of Life Sciences, University of Sussex, Falmer, Brighton, East Sussex, BN1 9QG, UK
| | - Luca Biasetti
- School of Life Sciences, University of Sussex, Falmer, Brighton, East Sussex, BN1 9QG, UK
| | - Saskia J Pollack
- School of Life Sciences, University of Sussex, Falmer, Brighton, East Sussex, BN1 9QG, UK
| | - James P Quinn
- School of Life Sciences, University of Sussex, Falmer, Brighton, East Sussex, BN1 9QG, UK
| | - Julian R Thorpe
- School of Life Sciences, University of Sussex, Falmer, Brighton, East Sussex, BN1 9QG, UK
| | - Aidan J Doherty
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, BN1 9RQ, UK
| | - Louise C Serpell
- School of Life Sciences, University of Sussex, Falmer, Brighton, East Sussex, BN1 9QG, UK.
| |
Collapse
|
33
|
Abstract
The nucleolus is a prominent subnuclear compartment, where ribosome biosynthesis takes place. Recently, the nucleolus has gained attention for its novel role in the regulation of cellular stress. Nucleolar stress is emerging as a new concept, which is characterized by diverse cellular insult-induced abnormalities in nucleolar structure and function, ultimately leading to activation of p53 or other stress signaling pathways and alterations in cell behavior. Despite a number of comprehensive reviews on this concept, straightforward and clear-cut way criteria for a nucleolar stress state, regarding the factors that elicit this state, the morphological and functional alterations as well as the rationale for p53 activation are still missing. Based on literature of the past two decades, we herein summarize the evolution of the concept and provide hallmarks of nucleolar stress. Along with updated information and thorough discussion of existing confusions in the field, we pay particular attention to the current understanding of the sensing mechanisms, i.e., how stress is integrated by p53. In addition, we propose our own emphasis regarding the role of nucleolar protein NPM1 in the hallmarks of nucleolar stress and sensing mechanisms. Finally, the links of nucleolar stress to human diseases are briefly and selectively introduced.
Collapse
Affiliation(s)
- Kai Yang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.,Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jie Yang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Jing Yi
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
34
|
Zhang Q, Chen ZS, An Y, Liu H, Hou Y, Li W, Lau KF, Koon AC, Ngo JCK, Chan HYE. A peptidylic inhibitor for neutralizing expanded CAG RNA-induced nucleolar stress in polyglutamine diseases. RNA (NEW YORK, N.Y.) 2018; 24:486-498. [PMID: 29295891 PMCID: PMC5855950 DOI: 10.1261/rna.062703.117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/27/2017] [Indexed: 06/07/2023]
Abstract
Polyglutamine (polyQ) diseases are a class of progressive neurodegenerative disorders characterized by the expression of both expanded CAG RNA and misfolded polyQ protein. We previously reported that the direct interaction between expanded CAG RNA and nucleolar protein nucleolin (NCL) impedes preribosomal RNA (pre-rRNA) transcription, and eventually triggers nucleolar stress-induced apoptosis in polyQ diseases. Here, we report that a 21-amino acid peptide, named "beta-structured inhibitor for neurodegenerative diseases" (BIND), effectively suppresses toxicity induced by expanded CAG RNA. When administered to a cell model, BIND potently inhibited cell death induced by expanded CAG RNA with an IC50 value of ∼0.7 µM. We showed that the function of BIND is dependent on Glu2, Lys13, Gly14, Ile18, Glu19, and Phe20. BIND treatment restored the subcellular localization of nucleolar marker protein and the expression level of pre-45s rRNA Through isothermal titration calorimetry analysis, we demonstrated that BIND suppresses nucleolar stress via a direct interaction with CAG RNA in a length-dependent manner. The mean binding constants (KD) of BIND to SCA2CAG22 , SCA2CAG42 , SCA2CAG55 , and SCA2CAG72 RNA are 17.28, 5.60, 4.83, and 0.66 µM, respectively. In vivo, BIND ameliorates retinal degeneration and climbing defects, and extends the lifespan of Drosophila expressing expanded CAG RNA. These effects suggested that BIND can suppress neurodegeneration in diverse polyQ disease models in vivo and in vitro without exerting observable cytotoxic effect. Our results collectively demonstrated that BIND is an effective inhibitor of expanded CAG RNA-induced toxicity in polyQ diseases.
Collapse
Affiliation(s)
- Qian Zhang
- Laboratory of Drosophila Research, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Zhefan Stephen Chen
- Laboratory of Drosophila Research, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Ying An
- Laboratory of Drosophila Research, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Haizhen Liu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Yonghui Hou
- Laboratory of Drosophila Research, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Wen Li
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Kwok-Fai Lau
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Alex Chun Koon
- Laboratory of Drosophila Research, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Jacky Chi Ki Ngo
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Ho Yin Edwin Chan
- Laboratory of Drosophila Research, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
- Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| |
Collapse
|
35
|
mTOR: An attractive therapeutic target for osteosarcoma? Oncotarget 2018; 7:50805-50813. [PMID: 27177330 PMCID: PMC5226621 DOI: 10.18632/oncotarget.9305] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/05/2016] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma (OS) is a common primary malignant bone tumor with high morbidity and mortality in children and young adults. How to improve poor prognosis of OS due to resistance to chemotherapy remains a challenge. Recently, growing findings show activation of mammalian target of rapamycin (mTOR), is associated with OS cell growth, proliferation, metastasis. Targeting mTOR may be a promising therapeutic approach for treating OS. This review summarizes the roles of mTOR pathway in OS and present research status of mTOR inhibitors in the context of OS. In addition, we have attempted to discuss how to design a better treatment project for OS by combining mTOR inhibitor with other drugs.
Collapse
|
36
|
Farhane Z, Bonnier F, Howe O, Casey A, Byrne HJ. Doxorubicin kinetics and effects on lung cancer cell lines using in vitro Raman micro-spectroscopy: binding signatures, drug resistance and DNA repair. JOURNAL OF BIOPHOTONICS 2018. [PMID: 28635172 DOI: 10.1002/jbio.201700060] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Raman micro-spectroscopy is a non-invasive analytical tool, whose potential in cellular analysis and monitoring drug mechanisms of action has already been demonstrated, and which can potentially be used in pre-clinical and clinical applications for the prediction of chemotherapeutic efficacy. To further investigate such potential clinical application, it is important to demonstrate its capability to differentiate drug mechanisms of action and cellular resistances. Using the example of Doxorubicin (DOX), in this study, it was used to probe the cellular uptake, signatures of chemical binding and subsequent cellular responses, of the chemotherapeutic drug in two lung cancer cell lines, A549 and Calu-1. Multivariate statistical analysis was used to elucidate the spectroscopic signatures associated with DOX uptake and subcellular interaction. Biomarkers related to DNA damage and repair, and mechanisms leading to apoptosis were also measured and correlated to Raman spectral profiles. Results confirm the potential of Raman spectroscopic profiling to elucidate both drug kinetics and pharmacodynamics and differentiate cellular drug resistance associated with different subcellular accumulation rates and subsequent cellular response to DNA damage, pointing towards a better understanding of drug resistance for personalised targeted treatment.
Collapse
Affiliation(s)
- Zeineb Farhane
- FOCAS Research Institute, Dublin Institute of Technology, Kevin Street, Dublin 8, Ireland
- School of Physics, Dublin Institute of Technology, Kevin Street, Dublin 8, Ireland
| | - Franck Bonnier
- Université François-Rabelais de Tours, Faculty of Pharmacy, EA 6295 Nanomédicaments et Nanosondes, 31 avenue Monge, 37200 Tours, France
| | - Orla Howe
- School of Biological Sciences, Dublin Institute of Technology, Kevin Street, Dublin 8, Ireland
| | - Alan Casey
- FOCAS Research Institute, Dublin Institute of Technology, Kevin Street, Dublin 8, Ireland
| | - Hugh J Byrne
- FOCAS Research Institute, Dublin Institute of Technology, Kevin Street, Dublin 8, Ireland
| |
Collapse
|
37
|
Mo Y, Lin R, Liu P, Tan M, Xiong Y, Guan KL, Yuan HX. SIRT7 deacetylates DDB1 and suppresses the activity of the CRL4 E3 ligase complexes. FEBS J 2017; 284:3619-3636. [PMID: 28886238 DOI: 10.1111/febs.14259] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/28/2017] [Accepted: 08/31/2017] [Indexed: 12/14/2022]
Abstract
Cullin 4 (CUL4) and small ring finger protein ROC1 assemble to form E3 ubiquitin ligase (CRL4) complexes. CUL4 interacts with WD-40 proteins through the adaptor protein DNA damage-binding protein 1 (DDB1) to target substrates for ubiquitylation. Very little is known on how the CUL4 and DDB1 interaction is regulated. Here, we show that DDB1 is acetylated and acetylation promotes DDB1 binding to CUL4. We also identify nucleolar sirtuin 7 (SIRT7) as a major deacetylase that negatively regulates DDB1-CUL4 interaction. Following inhibition of nucleolar function by actinomycin D or 5-fluorouracil treatment or knocking down the gene for the RNA polymerase I component UBF, SIRT7 is mobilized from the nucleolus to the nucleoplasm and promotes DDB1 deacetylation, leading to decreased DDB1-CUL4 association and CRL4 activity. This results in the accumulation or activation of CRL4 substrates including LATS1 and p73, which contribute to cell apoptosis induced by actinomycin D and 5-fluorouracil. Our study uncovers a novel regulation of CRL4 E3 ligase complexes.
Collapse
Affiliation(s)
- Yan Mo
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ran Lin
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Peng Liu
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Minjia Tan
- The Chemical Proteomics Center and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yue Xiong
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Lineberger Comprehensive Cancer Center, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, NC, USA
| | - Kun-Liang Guan
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Hai-Xin Yuan
- The Fifth People's Hospital of Shanghai and the Molecular and Cell Biology Research Lab of the Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Zhao S, Xia Y, Zhang F, Xiong Z, Li Y, Yan W, Chen X, Wang W, Wang H, Gao E, Lee Y, Li C, Wang S, Zhang L, Tao L. Nucleostemin dysregulation contributes to ischemic vulnerability of diabetic hearts: Role of ribosomal biogenesis. J Mol Cell Cardiol 2017; 108:106-113. [PMID: 28549781 DOI: 10.1016/j.yjmcc.2017.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/29/2017] [Accepted: 05/22/2017] [Indexed: 01/20/2023]
Abstract
Diabetes is a major health problem worldwide. As well-known, diabetes greatly increases cardiac vulnerability to ischemia/reperfusion (I/R) injury, but the underlying mechanisms remain elusive. Nucleostemin (NS) is a nucleolar protein that controls ribosomal biogenesis and exerts cardioprotective effects against I/R injury. However, whether NS-mediated ribosomal biogenesis regulates ischemic vulnerability of diabetic hearts remains unanswered. Utilizing myocardial I/R mouse models, we found that cardiac NS expression significantly increased in response to I/R in normal diet (ND)-fed mice. Surprisingly, cardiac NS failed to be upregulated in high fat diet (HFD)-induced diabetic mice, accompanied by obvious ribosomal dysfunction. Compared with ND group, cardiac specific overexpression of NS by adenovirus (AV) injection significantly restored I/R-induced ribosomal function enhancement, reduced cardiomyocyte apoptosis, improved cardiac function, and decreased infarct sizes in diabetic mice. Notably, co-treatment of homoharringtonine (HHT), a selective inhibitor of ribosomal function, totally blocked NS-mediated cardioprotective effects against I/R injury. Furthermore, in cultured cardiomyocytes, saturated fatty acids treatment, but not high glucose exposure, significantly inhibited simulated I/R-induced NS upregulation and ribosomal function improvement. In conclusion, these data for the first time demonstrate that NS dysregulation induced by saturated fatty acids exposure might be an important cause of increased ischemic vulnerability to I/R injury in diabetic hearts. Targeting NS dysregulation and subsequent ribosomal dysfunction could be a promising therapeutic strategy for diabetic I/R injury management.
Collapse
Affiliation(s)
- Shihao Zhao
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Yunlong Xia
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China; Department of Physiology, the Fourth Military Medical University, China; Department of Cardiology, the 201st Hospital of People's Liberation Army, China
| | - Zhenyu Xiong
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Yueyang Li
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Xiyao Chen
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, China
| | - Wei Wang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Helin Wang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Erhe Gao
- Center for Translational Medicine, Temple University, United States
| | - Yan Lee
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Shan Wang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Ling Zhang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, China.
| |
Collapse
|
39
|
Popov LD. Mitochondrial networking in diabetic left ventricle cardiomyocytes. Mitochondrion 2016; 34:24-31. [PMID: 28007605 DOI: 10.1016/j.mito.2016.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 10/07/2016] [Accepted: 12/16/2016] [Indexed: 12/17/2022]
Abstract
Cardiomyocyte mitochondria preserve "the quorum sensing" attribute of their aerobic bacterial ancestors, as shown by the transient physical connectivity and communication not only with each other, but also with other intracellular organelles and with cytosol, ensuing cellular homeostasis. In this review, we present original electron microscopy evidence on mitochondrial networking within diabetic left ventricular cardiomyocytes, focusing on: (i) the inter-mitochondrial communication, allowing electrochemical signals transfer and outer membrane components or matrix proteins exchange, (ii) the interplay between mitochondria and the cardiomyocyte nucleus, nucleolus, sarcoplasmic reticulum, lysosomes, and lipid droplets viewed as attributes of mitochondrial "quality control" and "retrograde signaling function", and (iii) the crosstalk between mitochondria and cardiomyocyte cytosol, as part of the adaptive responses that allow cells survival. Confirmation of such interactions in diabetic myocardium and identification of molecules involved are ongoing, foreseeing the alleviation of heart contractile dysfunction in cardiomyopathy.
Collapse
Affiliation(s)
- Lucia-Doina Popov
- "Nicolae Simionescu" Institute of Cellular Biology and Pathology of the Romanian Academy, 8, B.P. Hasdeu Street, Bucharest 050568, Romania.
| |
Collapse
|
40
|
Kung ML, Hsieh CW, Tai MH, Weng CH, Wu DC, Wu WJ, Yeh BW, Hsieh SL, Kuo CH, Hung HS, Hsieh S. Nanoscale characterization illustrates the cisplatin-mediated biomechanical changes of B16-F10 melanoma cells. Phys Chem Chem Phys 2016; 18:7124-31. [PMID: 26886764 DOI: 10.1039/c5cp07971c] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cells reorganize their membrane biomechanical dynamics in response to environmental stimuli or inhibitors associated with their physiological/pathological processes, and disease therapeutics. To validate the biophysical dynamics during cell exposure to anti-cancer drugs, we investigate the nanoscale biological characterization in melanoma cells undergoing cisplatin treatment. Using atomic force microscopy, we demonstrate that the cellular morphology and membrane ultrastructure are altered after exposure to cisplatin. In contrast to their normal spindle-like shape, cisplatin causes cell deformation rendering cells flat and enlarged, which increases the cell area by 3-4 fold. Additionally, cisplatin decreases the topography height values for both the cytoplasmic and nuclear regions (by 40-80% and 60%, respectively). Furthermore, cisplatin increases the cytoplasmic root mean square roughness by 110-240% in correlation with the drug concentration and attenuates the nuclear RMS by 60%. Moreover, the cellular adhesion force was enhanced, while the Young's modulus elasticity was attenuated by ∼2 and ∼2.3 fold, respectively. F-actin phalloidin staining revealed that cisplatin enlarges the cell size through enhanced stress fiber formation and promotes cytoskeletal reorganization. Immunoblot analyses further revealed that the activities of focal adhesion proteins, such as FAK and c-Src, are upregulated by cisplatin through phosphorylation at tyrosine 397 and 530, respectively. Collectively, these results show that cisplatin-treated melanoma cells not only exhibit the upregulation of FAK-mediated signaling to enhance the cytoskeleton mechanical stretch, but also promote the cytoskeletal rearrangement resulting in 43% decrease in the cell modulus. These mechanisms thus promote the malignancy and invasiveness of the melanoma cells.
Collapse
Affiliation(s)
- Mei-Lang Kung
- Department of Chemistry, National Sun Yat-sen University, 70 Lien-hai Rd., Kaohsiung 80424, Taiwan.
| | - Chiung-Wen Hsieh
- Department of Chemistry, National Sun Yat-sen University, 70 Lien-hai Rd., Kaohsiung 80424, Taiwan.
| | - Ming-Hong Tai
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan and Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan and Center for Neuroscience, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan and Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chien-Hui Weng
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Deng-Chyang Wu
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan and Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80708, Taiwan
| | - Wen-Jeng Wu
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Bi-Wen Yeh
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Shu-Ling Hsieh
- Department of Seafood Science, National Kaohsiung Marine University, Kaohsiung 80811, Taiwan
| | - Chao-Hung Kuo
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan and Department of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Huey-Shan Hung
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan and Center for Neuropsychiatry, China Medical University Hospital, Taichung 40402, Taiwan
| | - Shuchen Hsieh
- Department of Chemistry, National Sun Yat-sen University, 70 Lien-hai Rd., Kaohsiung 80424, Taiwan. and Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan and School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
41
|
Musinova YR, Lisitsyna OM, Sorokin DV, Arifulin EA, Smirnova TA, Zinovkin RA, Potashnikova DM, Vassetzky YS, Sheval EV. RNA-dependent disassembly of nuclear bodies. J Cell Sci 2016; 129:4509-4520. [PMID: 27875271 DOI: 10.1242/jcs.189142] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 11/02/2016] [Indexed: 12/17/2022] Open
Abstract
Nuclear bodies are membraneless organelles that play important roles in genome functioning. A specific type of nuclear bodies known as interphase prenucleolar bodies (iPNBs) are formed in the nucleoplasm after hypotonic stress from partially disassembled nucleoli. iPNBs are then disassembled, and the nucleoli are reformed simultaneously. Here, we show that diffusion of B23 molecules (also known as nucleophosmin, NPM1) from iPNBs, but not fusion of iPNBs with the nucleoli, contributes to the transfer of B23 from iPNBs to the nucleoli. Maturation of pre-ribosomal RNAs (rRNAs) and the subsequent outflow of mature rRNAs from iPNBs led to the disassembly of iPNBs. We found that B23 transfer was dependent on the synthesis of pre-rRNA molecules in nucleoli; these pre-rRNA molecules interacted with B23 and led to its accumulation within nucleoli. The transfer of B23 between iPNBs and nucleoli was accomplished through a nucleoplasmic pool of B23, and increased nucleoplasmic B23 content retarded disassembly, whereas B23 depletion accelerated disassembly. Our results suggest that iPNB disassembly and nucleolus assembly might be coupled through RNA-dependent exchange of nucleolar proteins, creating a highly dynamic system with long-distance correlations between spatially distinct processes.
Collapse
Affiliation(s)
- Yana R Musinova
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow 119992, Russia.,LIA1066 French-Russian Joint Cancer Research Laboratory, Villejuif 94805, France
| | - Olga M Lisitsyna
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow 119992, Russia
| | - Dmitry V Sorokin
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Botanická 68a, Brno 602 00, Czech Republic.,Laboratory of Mathematical Methods of Image Processing, Faculty of Computational Mathematics and Cybernetics, M.V. Lomonosov Moscow State University, Moscow 119992, Russia
| | - Eugene A Arifulin
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow 119992, Russia
| | - Tatiana A Smirnova
- Department of Cell Biology and Histology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow 119992, Russia
| | - Roman A Zinovkin
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow 119992, Russia
| | - Daria M Potashnikova
- Department of Cell Biology and Histology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow 119992, Russia
| | - Yegor S Vassetzky
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow 119992, Russia.,LIA1066 French-Russian Joint Cancer Research Laboratory, Villejuif 94805, France.,UMR8126, Université Paris-Sud, CNRS, Institut de cancérologie Gustave Roussy, Villejuif 94805, France
| | - Eugene V Sheval
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow 119992, Russia .,LIA1066 French-Russian Joint Cancer Research Laboratory, Villejuif 94805, France
| |
Collapse
|
42
|
Abstract
Noncoding RNAs are an emerging class of nonpeptide regulators of metabolism. Metabolic diseases and the altered metabolic environment induce marked changes in levels of microRNAs and long noncoding RNAs. Furthermore, recent studies indicate that a growing number of microRNAs and long noncoding RNAs serve as critical mediators of adaptive and maladaptive responses through their effects on gene expression. The metabolic environment also has a profound impact on the functions of classes of noncoding RNAs that have been thought primarily to subserve housekeeping functions in cells-ribosomal RNAs, transfer RNAs, and small nucleolar RNAs. Evidence is accumulating that these RNAs are also components of an integrated cellular response to the metabolic milieu. This Perspective discusses the different classes of noncoding RNAs and their contributions to the pathogenesis of metabolic stress.
Collapse
Affiliation(s)
- George Caputa
- Diabetic Cardiovascular Disease Center and Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Jean E Schaffer
- Diabetic Cardiovascular Disease Center and Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| |
Collapse
|
43
|
Hernández-Ortega K, Garcia-Esparcia P, Gil L, Lucas JJ, Ferrer I. Altered Machinery of Protein Synthesis in Alzheimer's: From the Nucleolus to the Ribosome. Brain Pathol 2015; 26:593-605. [PMID: 26512942 DOI: 10.1111/bpa.12335] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 10/22/2015] [Indexed: 12/17/2022] Open
Abstract
Ribosomes and protein synthesis have been reported to be altered in the cerebral cortex at advanced stages of Alzheimer's disease (AD). Modifications in the hippocampus with disease progression have not been assessed. Sixty-seven cases including middle-aged (MA) and AD stages I-VI were analyzed. Nucleolar chaperones nucleolin, nucleophosmin and nucleoplasmin 3, and upstream binding transcription factor RNA polymerase I gene (UBTF) mRNAs are abnormally regulated and their protein levels reduced in AD. Histone modifications dimethylated histone H3K9 (H3K9me2) and acetylated histone H3K12 (H3K12ac) are decreased in CA1. Nuclear tau declines in CA1 and dentate gyrus (DG), and practically disappears in neurons with neurofibrillary tangles. Subunit 28 ribosomal RNA (28S rRNA) expression is altered in CA1 and DG in AD. Several genes encoding ribosomal proteins are abnormally regulated and protein levels of translation initiation factors eIF2α, eIF3η and eIF5, and elongation factor eEF2, are altered in the CA1 region in AD. These findings show alterations in the protein synthesis machinery in AD involving the nucleolus, nucleus and ribosomes in the hippocampus in AD some of them starting at first stages (I-II) preceding neuron loss. These changes may lie behind reduced numbers of dendritic branches and reduced synapses of CA1 and DG neurons which cause hippocampal atrophy.
Collapse
Affiliation(s)
- Karina Hernández-Ortega
- Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Bellvitge University Hospital, University of Barcelona, Hospitalet de Llobregat, Spain.,Neuropathology, CIBERNED (Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas), Madrid, Spain
| | - Paula Garcia-Esparcia
- Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Bellvitge University Hospital, University of Barcelona, Hospitalet de Llobregat, Spain.,Neuropathology, CIBERNED (Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas), Madrid, Spain
| | - Laura Gil
- Department of Genetics, Medical School, Alfonso X el Sabio University (UAX), Villanueva de la Cañada; Centro de Investigaciones Biologicas (CIB), CSIC, Madrid, Spain
| | - José J Lucas
- Neuropathology, CIBERNED (Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas), Madrid, Spain.,Department of Molecular Biology, Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain
| | - Isidre Ferrer
- Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Bellvitge University Hospital, University of Barcelona, Hospitalet de Llobregat, Spain.,Neuropathology, CIBERNED (Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas), Madrid, Spain
| |
Collapse
|
44
|
Garcia-Esparcia P, Hernández-Ortega K, Koneti A, Gil L, Delgado-Morales R, Castaño E, Carmona M, Ferrer I. Altered machinery of protein synthesis is region- and stage-dependent and is associated with α-synuclein oligomers in Parkinson's disease. Acta Neuropathol Commun 2015; 3:76. [PMID: 26621506 PMCID: PMC4666041 DOI: 10.1186/s40478-015-0257-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/14/2015] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Parkinson's disease (PD) is characterized by the accumulation of abnormal α-synuclein in selected regions of the brain following a gradient of severity with disease progression. Whether this is accompanied by globally altered protein synthesis is poorly documented. The present study was carried out in PD stages 1-6 of Braak and middle-aged (MA) individuals without alterations in brain in the substantia nigra, frontal cortex area 8, angular gyrus, precuneus and putamen. RESULTS Reduced mRNA expression of nucleolar proteins nucleolin (NCL), nucleophosmin (NPM1), nucleoplasmin 3 (NPM3) and upstream binding transcription factor (UBF), decreased NPM1 but not NPM3 nucleolar protein immunostaining in remaining neurons; diminished 18S rRNA, 28S rRNA; reduced expression of several mRNAs encoding ribosomal protein (RP) subunits; and altered protein levels of initiation factor eIF3 and elongation factor eEF2 of protein synthesis was found in the substantia nigra in PD along with disease progression. Although many of these changes can be related to neuron loss in the substantia nigra, selective alteration of certain factors indicates variable degree of vulnerability of mRNAs, rRNAs and proteins in degenerating sustantia nigra. NPM1 mRNA and 18S rRNA was increased in the frontal cortex area 8 at stage 5-6; modifications were less marked and region-dependent in the angular gyrus and precuneus. Several RPs were abnormally regulated in the frontal cortex area 8 and precuneus, but only one RP in the angular gyrus, in PD. Altered levels of eIF3 and eIF1, and decrease eEF1A and eEF2 protein levels were observed in the frontal cortex in PD. No modifications were found in the putamen at any time of the study except transient modifications in 28S rRNA and only one RP mRNA at stages 5-6. These observations further indicate marked region-dependent and stage-dependent alterations in the cerebral cortex in PD. Altered solubility and α-synuclein oligomer formation, assessed in total homogenate fractions blotted with anti-α-synuclein oligomer-specific antibody, was demonstrated in the substantia nigra and frontal cortex, but not in the putamen, in PD. Dramatic increase in α-synuclein oligomers was also seen in fluorescent-activated cell sorter (FACS)-isolated nuclei in the frontal cortex in PD. CONCLUSIONS Altered machinery of protein synthesis is altered in the substantia nigra and cerebral cortex in PD being the frontal cortex area 8 more affected than the angular gyrus and precuneus; in contrast, pathways of protein synthesis are apparently preserved in the putamen. This is associated with the presence of α-synuclein oligomeric species in total homogenates; substantia nigra and frontal cortex are enriched, albeit with different band patterns, in α-synuclein oligomeric species, whereas α-synuclein oligomers are not detected in the putamen.
Collapse
Affiliation(s)
- Paula Garcia-Esparcia
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Karina Hernández-Ortega
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Anusha Koneti
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Laura Gil
- Department of Genetics, Medical School, Alfonso X el Sabio University, Villanueva de la Cañada, Madrid, Spain
| | - Raul Delgado-Morales
- Cancer Epigenetics and Biology Program, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Ester Castaño
- Biology-Bellvitge Unit, Scientific and Technological Centers-University of Barcelona (CCiTUB), Hospitalet de Llobregat, Barcelona, Spain
| | - Margarita Carmona
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Isidre Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.
- Institute of Neuropathology, Service of Pathologic Anatomy, Bellvitge University Hospital, carrer Feixa Llarga s/n, 08907, Hospitalet de Llobregat, Spain.
| |
Collapse
|
45
|
RRP12 is a crucial nucleolar protein that regulates p53 activity in osteosarcoma cells. Tumour Biol 2015; 37:4351-8. [PMID: 26499779 DOI: 10.1007/s13277-015-4062-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/04/2015] [Indexed: 10/22/2022] Open
Abstract
RRP12 (ribosomal RNA processing 12 homolog), a nucleolar protein, plays important roles in cell cycle progression and the response to deoxyribonucleic acid (DNA) damage in yeast cells. However, its role has not been investigated in mammalian cells that possess p53, which has close functional association to nucleolus. We explored the role of RRP12 in nucleolar stress condition using an osteosarcoma cell line, U2OS. To induce DNA damage and nucleolar disruption, two cytotoxic drugs, doxorubicin and actinomycin D were used. Cytotoxic stress resulted nucleolar disruption induced cell cycle arrest and apoptosis in U2OS cells. However, RRP12 overexpression promoted resistance to cytotoxic stress. In contrast, RRP12 silencing enhanced susceptibility to cytotoxic stress. During drug treatment, p53 activity and cell death were suppressed by RRP12 overexpression but promoted by RRP12 silencing. This study demonstrated that RRP12 was crucial for cell survival during cytotoxic stress via the repression of p53 stability. Thus, targeting RRP12 may enhance chemotherapeutic effect in cancers.
Collapse
|
46
|
Goudarzi KM, Nistér M, Lindström MS. mTOR inhibitors blunt the p53 response to nucleolar stress by regulating RPL11 and MDM2 levels. Cancer Biol Ther 2015; 15:1499-514. [PMID: 25482947 DOI: 10.4161/15384047.2014.955743] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a master regulator of cell growth through its ability to stimulate ribosome biogenesis and mRNA translation. In contrast, the p53 tumor suppressor negatively controls cell growth and is activated by a wide range of insults to the cell. The mTOR and p53 signaling pathways are connected by a number of different mechanisms. Chemotherapeutics that inhibit ribosome biogenesis often induce nucleolar stress and activation of p53. Here we have investigated how the p53 response to nucleolar stress is affected by simultaneous mTOR inhibition in osteosarcoma and glioma cell lines. We found that inhibitors of the mTOR pathway including rapamycin, wortmannin, and caffeine blunted the p53 response to nucleolar stress induced by actinomycin D. Synthetic inhibitors of mTOR (temsirolimus, LY294.002 and PP242) also impaired actinomycin D triggered p53 stabilization and induction of p21. Ribosomal protein (RPL11) is known to be required for p53 protein stabilization following nucleolar stress. Treatment of cells with mTOR inhibitors may lead to reduced synthesis of RPL11 and thereby destabilize p53. We found that rapamycin mimicked the effect of RPL11 depletion in terms of blunting the p53 response to nucleolar stress. However, the extent to which the levels of p53 and RPL11 were reduced by rapamycin varied between cell lines. Additional mechanisms whereby rapamycin blunts the p53 response to nucleolar stress are likely to be involved. Indeed, rapamycin increased the levels of endogenous MDM2 despite inhibition of its phosphorylation at Ser-166. Our findings may have implications for the design of combinatorial cancer treatments with mTOR pathway inhibitors.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- Act D, actinomycin D
- BrdU, bromodeoxyuridine
- CHX, cycloheximide
- DMSO, dimethylsulphoxide
- DOX, doxorubicin
- EGCG, epigallocatechin-3-gallate
- FACS, fluorescence-activated cell sorting
- MPA, mycophenolic acid
- MTT, (3-[4, 5-dimethylthiazol-2-yl]-2, 5 diphenyl tetrazolium bromide)
- PI, propidium iodide
- actinomycin D
- caffeine
- glioma
- mTOR
- mTOR, mechanistic target of rapamycin
- nutlin-3
- p21
- p53
- rapamycin
- ribosomal protein L11
- ribosome biogenesis
Collapse
Affiliation(s)
- Kaveh M Goudarzi
- a Department of Oncology-Pathology; Karolinska Institutet; Cancer Center Karolinska ; Karolinska University Hospital ; Stockholm , Sweden
| | | | | |
Collapse
|
47
|
Abstract
A veritable explosion of primary research papers within the past 10 years focuses on nucleolar and ribosomal stress, and for good reason: with ribosome biosynthesis consuming ~80% of a cell’s energy, nearly all metabolic and signaling pathways lead ultimately to or from the nucleolus. We begin by describing p53 activation upon nucleolar stress resulting in cell cycle arrest or apoptosis. The significance of this mechanism cannot be understated, as oncologists are now inducing nucleolar stress strategically in cancer cells as a potential anti-cancer therapy. We also summarize the human ribosomopathies, syndromes in which ribosome biogenesis or function are impaired leading to birth defects or bone narrow failures; the perplexing problem in the ribosomopathies is why only certain cells are affected despite the fact that the causative mutation is systemic. We then describe p53-independent nucleolar stress, first in yeast which lacks p53, and then in other model metazoans that lack MDM2, the critical E3 ubiquitin ligase that normally inactivates p53. Do these presumably ancient p53-independent nucleolar stress pathways remain latent in human cells? If they still exist, can we use them to target >50% of known human cancers that lack functional p53?
Collapse
Affiliation(s)
- Allison James
- a Department of Biological Sciences; Louisiana State University; Baton Rouge, LA USA
| | | | | | | | | |
Collapse
|
48
|
Ranieri D, Avitabile D, Shiota M, Yokomizo A, Naito S, Bizzarri M, Torrisi MR. Nuclear redox imbalance affects circadian oscillation in HaCaT keratinocytes. Int J Biochem Cell Biol 2015; 65:113-24. [PMID: 26028291 DOI: 10.1016/j.biocel.2015.05.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 04/30/2015] [Accepted: 05/15/2015] [Indexed: 12/29/2022]
Abstract
Circadian clock is regulated by a transcriptional/translational feedback loop (TTFL) lasting ∼24 h. Circadian oscillation of peroxiredoxins (PRDX1-6) redox status has been shown in mature erythrocytes. We have recently reported that nuclear levels of PRDX2 are circadian regulated in the HaCaT keratinocytes. In this study, we addressed whether PRDX2 translocation could influence the TTFL. A reporter HaCaT cell line stably expressing the luciferase gene under control of Bmal1 promoter was lentivirally transduced either with an empty vector (EV), a vector carrying a myc-tagged wild type PRDX2 (PRDX2-Myc) or the same gene with a nuclear localization sequence (PRDX2-MycNuc). PRDX2 overexpressing cells were protected from H2O2-induced oxidative stress. The amplitude of the Bmal1 promoter activity was significantly dampened in PRDX2-MycNuc versus EV cells when synchronized either by dexamethasone treatment or temperature cycles. Clock synchronization was not affected in PRDX2 silenced cells. N-acetyl cysteine or melatonin treatments, significantly dampened the Bmal1 promoter activity suggesting that sustained scavenging of ROS impairs clock synchronization. Noteworthy, H2O2 treatment rescued proper oscillation of the clock in synchronized PRDX2-MycNuc HaCaT cells. Since the histone deacetylase Sirtuin 1 (Sirt1) modulates clock gene expression amplitude, the effect of Sirt1 activator resveratrol or Sirt1 inhibitor nicotinamide were also investigated. Interestingly, NAM enhanced the molecular clock synchronization in PRDX2-MycNuc cells. Our findings demonstrate that PRDX2 regulates the TTFL oscillation by finely tuning the cellular redox status of the nucleus likely influencing the deacetilase activity of SIRT1 enzyme.
Collapse
Affiliation(s)
- Danilo Ranieri
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Daniele Avitabile
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy; Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, Milan, Italy.
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Yokomizo
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiji Naito
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mariano Bizzarri
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Italy
| | - Maria Rosaria Torrisi
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy; Azienda Ospedaliera S. Andrea, Rome, Italy
| |
Collapse
|
49
|
Holley CL, Li MW, Scruggs BS, Matkovich SJ, Ory DS, Schaffer JE. Cytosolic accumulation of small nucleolar RNAs (snoRNAs) is dynamically regulated by NADPH oxidase. J Biol Chem 2015; 290:11741-8. [PMID: 25792744 DOI: 10.1074/jbc.m115.637413] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Indexed: 01/22/2023] Open
Abstract
Small nucleolar RNAs (snoRNAs) guide nucleotide modifications of cellular RNAs in the nucleus. We previously showed that box C/D snoRNAs from the Rpl13a locus are unexpected mediators of physiologic oxidative stress, independent of their predicted ribosomal RNA modifications. Here we demonstrate that oxidative stress induced by doxorubicin causes rapid cytoplasmic accumulation of the Rpl13a snoRNAs through a mechanism that requires superoxide and a nuclear splice variant of NADPH oxidase. RNA-sequencing analysis reveals that box C/D snoRNAs as a class are present in the cytoplasm, where their levels are dynamically regulated by NADPH oxidase. These findings suggest that snoRNAs may orchestrate the response to environmental stress through molecular interactions outside of the nucleus.
Collapse
Affiliation(s)
| | - Melissa W Li
- From the Diabetic Cardiovascular Disease Center and
| | | | - Scot J Matkovich
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missiouri 63110
| | - Daniel S Ory
- From the Diabetic Cardiovascular Disease Center and
| | | |
Collapse
|
50
|
Hariharan N, Quijada P, Mohsin S, Joyo A, Samse K, Monsanto M, De La Torre A, Avitabile D, Ormachea L, McGregor MJ, Tsai EJ, Sussman MA. Nucleostemin rejuvenates cardiac progenitor cells and antagonizes myocardial aging. J Am Coll Cardiol 2015; 65:133-47. [PMID: 25593054 DOI: 10.1016/j.jacc.2014.09.086] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/03/2014] [Accepted: 09/23/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Functional decline in stem cell-mediated regeneration contributes to aging associated with cellular senescence in c-kit+ cardiac progenitor cells (CPCs). Clinical implementation of CPC-based therapy in elderly patients would benefit tremendously from understanding molecular characteristics of senescence to antagonize aging. Nucleostemin (NS) is a nucleolar protein regulating stem cell proliferation and pluripotency. OBJECTIVES This study sought to demonstrate that NS preserves characteristics associated with "stemness" in CPCs and antagonizes myocardial senescence and aging. METHODS CPCs isolated from human fetal (fetal human cardiac progenitor cell [FhCPC]) and adult failing (adult human cardiac progenitor cell [AhCPC]) hearts, as well as young (young cardiac progenitor cell [YCPC]) and old mice (old cardiac progenitor cell [OCPC]), were studied for senescence characteristics and NS expression. Heterozygous knockout mice with 1 functional allele of NS (NS+/-) were used to demonstrate that NS preserves myocardial structure and function and slows characteristics of aging. RESULTS NS expression is decreased in AhCPCs relative to FhCPCs, correlating with lowered proliferation potential and shortened telomere length. AhCPC characteristics resemble those of OCPCs, which have a phenotype induced by NS silencing, resulting in cell flattening, senescence, multinucleated cells, decreased S-phase progression, diminished expression of stemness markers, and up-regulation of p53 and p16. CPC senescence resulting from NS loss is partially p53 dependent and is rescued by concurrent silencing of p53. Mechanistically, NS induction correlates with Pim-1 kinase-mediated stabilization of c-Myc. Engineering OCPCs and AhCPCs to overexpress NS decreases senescent and multinucleated cells, restores morphology, and antagonizes senescence, thereby preserving phenotypic properties of "stemness." Early cardiac aging with a decline in cardiac function, an increase in senescence markers p53 and p16, telomere attrition, and accompanied CPC exhaustion is evident in NS+/- mice. CONCLUSIONS Youthful properties and antagonism of senescence in CPCs and the myocardium are consistent with a role for NS downstream from Pim-1 signaling that enhances cardiac regeneration.
Collapse
Affiliation(s)
- Nirmala Hariharan
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California
| | - Pearl Quijada
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California
| | - Sadia Mohsin
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California
| | - Anya Joyo
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California
| | - Kaitlen Samse
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California
| | - Megan Monsanto
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California
| | - Andrea De La Torre
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California
| | - Daniele Avitabile
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California; Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Lucia Ormachea
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California
| | - Michael J McGregor
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California
| | - Emily J Tsai
- Section in Cardiology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Mark A Sussman
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, California.
| |
Collapse
|