151
|
Dowling CM, Hollinshead KER, Di Grande A, Pritchard J, Zhang H, Dillon ET, Haley K, Papadopoulos E, Mehta AK, Bleach R, Lindner AU, Mooney B, Düssmann H, O'Connor D, Prehn JHM, Wynne K, Hemann M, Bradner JE, Kimmelman AC, Guerriero JL, Cagney G, Wong KK, Letai AG, Chonghaile TN. Multiple screening approaches reveal HDAC6 as a novel regulator of glycolytic metabolism in triple-negative breast cancer. SCIENCE ADVANCES 2021; 7:eabc4897. [PMID: 33523897 PMCID: PMC7810372 DOI: 10.1126/sciadv.abc4897] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/23/2020] [Indexed: 06/10/2023]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer without a targeted form of therapy. Unfortunately, up to 70% of patients with TNBC develop resistance to treatment. A known contributor to chemoresistance is dysfunctional mitochondrial apoptosis signaling. We set up a phenotypic small-molecule screen to reveal vulnerabilities in TNBC cells that were independent of mitochondrial apoptosis. Using a functional genetic approach, we identified that a "hit" compound, BAS-2, had a potentially similar mechanism of action to histone deacetylase inhibitors (HDAC). An in vitro HDAC inhibitor assay confirmed that the compound selectively inhibited HDAC6. Using state-of-the-art acetylome mass spectrometry, we identified glycolytic substrates of HDAC6 in TNBC cells. We confirmed that inhibition or knockout of HDAC6 reduced glycolytic metabolism both in vitro and in vivo. Through a series of unbiased screening approaches, we have identified a previously unidentified role for HDAC6 in regulating glycolytic metabolism.
Collapse
Affiliation(s)
- Catríona M Dowling
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Kate E R Hollinshead
- Department of Radiation Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Alessandra Di Grande
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Justin Pritchard
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Hua Zhang
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Eugene T Dillon
- School of Biomolecular and Biomedical Science, Conway Institute of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | - Kathryn Haley
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Eleni Papadopoulos
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Anita K Mehta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Rachel Bleach
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Andreas U Lindner
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Brian Mooney
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Heiko Düssmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Darran O'Connor
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kieran Wynne
- School of Biomolecular and Biomedical Science, Conway Institute of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | - Michael Hemann
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alec C Kimmelman
- Department of Radiation Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Jennifer L Guerriero
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Gerard Cagney
- School of Biomolecular and Biomedical Science, Conway Institute of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | - Kwok-Kin Wong
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Anthony G Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tríona Ní Chonghaile
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
152
|
The Molecular Mechanisms of Regulating Oxidative Stress-Induced Ferroptosis and Therapeutic Strategy in Tumors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8810785. [PMID: 33425217 PMCID: PMC7772020 DOI: 10.1155/2020/8810785] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/05/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022]
Abstract
Ferroptosis is an atypical form of regulated cell death, which is different from apoptosis, necrosis, pyroptosis, and autophagy. Ferroptosis is characterized by iron-dependent oxidative destruction of cellular membranes following the antioxidant system's failure. The sensitivity of ferroptosis is tightly regulated by a series of biological processes, the metabolism of iron, amino acids, and polyunsaturated fatty acids, and the interaction of glutathione (GSH), NADPH, coenzyme Q10 (CoQ10), and phospholipids. Elevated oxidative stress (ROS) level is a hallmark of cancer, and ferroptosis serves as a link between nutrition metabolism and redox biology. Targeting ferroptosis may be an effective and selective way for cancer therapy. The underlying molecular mechanism of ferroptosis occurrence is still not enough. This review will briefly summarize the process of ferroptosis and introduce critical molecules in the ferroptotic cascade. Furthermore, we reviewed the occurrence and regulation of reduction-oxidation (redox) for ferroptosis in cancer metabolism. The role of the tumor suppressor and the epigenetic regulator in tumor cell ferroptosis will also be described. Finally, old drugs that can be repurposed to induce ferroptosis will be characterized, aiming for drug repurposing and novel drug combinations for cancer therapy more efficiently and economically.
Collapse
|
153
|
Wang H, Wan X, Pilch PF, Ellisen LW, Fried SK, Liu L. An AMPK-dependent, non-canonical p53 pathway plays a key role in adipocyte metabolic reprogramming. eLife 2020; 9:63665. [PMID: 33320092 PMCID: PMC7758072 DOI: 10.7554/elife.63665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/14/2020] [Indexed: 12/27/2022] Open
Abstract
It has been known adipocytes increase p53 expression and activity in obesity, however, only canonical p53 functions (i.e. senescence and apoptosis) are attributed to inflammation-associated metabolic phenotypes. Whether or not p53 is directly involved in mature adipocyte metabolic regulation remains unclear. Here we show p53 protein expression can be up-regulated in adipocytes by nutrient starvation without activating cell senescence, apoptosis, or a death-related p53 canonical pathway. Inducing the loss of p53 in mature adipocytes significantly reprograms energy metabolism and this effect is primarily mediated through a AMP-activated protein kinase (AMPK) pathway and a novel downstream transcriptional target, lysosomal acid lipase (LAL). The pathophysiological relevance is further demonstrated in a conditional and adipocyte-specific p53 knockout mouse model. Overall, these data support a non-canonical p53 function in the regulation of adipocyte energy homeostasis and indicate that the dysregulation of this pathway may be involved in developing metabolic dysfunction in obesity.
Collapse
Affiliation(s)
- Hong Wang
- Departments of Pharmacology & Experimental Therapeutics, Boston University, School of Medicine, Boston, United States
| | - Xueping Wan
- Departments of Pharmacology & Experimental Therapeutics, Boston University, School of Medicine, Boston, United States
| | - Paul F Pilch
- Biochemistry, Boston University, School of Medicine, Boston, United States
| | - Leif W Ellisen
- Massachusetts General Hospital Cancer Center, Boston, United States.,Harvard Medical School, Boston, United States
| | - Susan K Fried
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Libin Liu
- Departments of Pharmacology & Experimental Therapeutics, Boston University, School of Medicine, Boston, United States
| |
Collapse
|
154
|
Glut1 expression is increased by p53 reduction to switch metabolism to glycolysis during osteoblast differentiation. Biochem J 2020; 477:1795-1811. [PMID: 32242617 DOI: 10.1042/bcj20190888] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022]
Abstract
The glycolytic system is selected for ATP synthesis not only in tumor cells but also in differentiated cells. Differentiated osteoblasts also switch the dominant metabolic pathway to aerobic glycolysis. We found that primary osteoblasts increased expressions of glycolysis-related enzymes such as Glut1, hexokinase 1 and 2, lactate dehydrogenase A and pyruvate kinase M2 during their differentiation. Osteoblast differentiation decreased expression of tumor suppressor p53, which negatively regulates Glut1 expression, and enhanced phosphorylation of AKT, which is regulated by phosphoinositol-3 kinase (PI3K). An inhibitor of PI3K enhanced p53 expression and repressed Glut1 expression. Luciferase reporter assay showed that p53 negatively regulated transcriptional activity of solute carrier family 2 member 1 gene promoter region. Inhibition of glycolysis in osteoblasts reduced ATP contents more significantly than inhibition of oxidative phosphorylation by carbonyl cyanide m-chlorophenyl hydrazine. These results have indicated that osteoblasts increase Glut1 expression through the down-regulation of p53 to switch their metabolic pathway to glycolysis during differentiation.
Collapse
|
155
|
Gebel J, Tuppi M, Sänger N, Schumacher B, Dötsch V. DNA Damaged Induced Cell Death in Oocytes. Molecules 2020; 25:molecules25235714. [PMID: 33287328 PMCID: PMC7730327 DOI: 10.3390/molecules25235714] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022] Open
Abstract
The production of haploid gametes through meiosis is central to the principle of sexual reproduction. The genetic diversity is further enhanced by exchange of genetic material between homologous chromosomes by the crossover mechanism. This mechanism not only requires correct pairing of homologous chromosomes but also efficient repair of the induced DNA double-strand breaks. Oocytes have evolved a unique quality control system that eliminates cells if chromosomes do not correctly align or if DNA repair is not possible. Central to this monitoring system that is conserved from nematodes and fruit fly to humans is the p53 protein family, and in vertebrates in particular p63. In mammals, oocytes are stored for a long time in the prophase of meiosis I which, in humans, can last more than 50 years. During the entire time of this arrest phase, the DNA damage checkpoint remains active. The treatment of female cancer patients with DNA damaging irradiation or chemotherapeutics activates this checkpoint and results in elimination of the oocyte pool causing premature menopause and infertility. Here, we review the molecular mechanisms of this quality control system and discuss potential therapeutic intervention for the preservation of the oocyte pool during chemotherapy.
Collapse
Affiliation(s)
- Jakob Gebel
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany; (J.G.); (M.T.)
| | - Marcel Tuppi
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany; (J.G.); (M.T.)
| | - Nicole Sänger
- Department for Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg-Campus 1, 53217 Bonn, Germany;
| | - Björn Schumacher
- Institute for Genome Stability in Aging and Disease, Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD) Research Center, and Center for Molecular Medicine, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany;
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany; (J.G.); (M.T.)
- Correspondence: ; Tel.: +49-69-798-29631
| |
Collapse
|
156
|
Oliveira GL, Coelho AR, Marques R, Oliveira PJ. Cancer cell metabolism: Rewiring the mitochondrial hub. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166016. [PMID: 33246010 DOI: 10.1016/j.bbadis.2020.166016] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/12/2020] [Accepted: 11/15/2020] [Indexed: 12/15/2022]
Abstract
To adapt to tumoral environment conditions or even to escape chemotherapy, cells rapidly reprogram their metabolism to handle adversities and survive. Given the rapid rise of studies uncovering novel insights and therapeutic opportunities based on the role of mitochondria in tumor metabolic programing and therapeutics, this review summarizes most significant developments in the field. Taking in mind the key role of mitochondria on carcinogenesis and tumor progression due to their involvement on tumor plasticity, metabolic remodeling, and signaling re-wiring, those organelles are also potential therapeutic targets. Among other topics, we address the recent data intersecting mitochondria as of prognostic value and staging in cancer, by mitochondrial DNA (mtDNA) determination, and current inhibitors developments targeting mtDNA, OXPHOS machinery and metabolic pathways. We contribute for a holistic view of the role of mitochondria metabolism and directed therapeutics to understand tumor metabolism, to circumvent therapy resistance, and to control tumor development.
Collapse
Affiliation(s)
- Gabriela L Oliveira
- CNC-Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal
| | - Ana R Coelho
- CNC-Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal
| | - Ricardo Marques
- CNC-Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal.
| |
Collapse
|
157
|
Masoud R, Reyes-Castellanos G, Lac S, Garcia J, Dou S, Shintu L, Abdel Hadi N, Gicquel T, El Kaoutari A, Diémé B, Tranchida F, Cormareche L, Borge L, Gayet O, Pasquier E, Dusetti N, Iovanna J, Carrier A. Targeting Mitochondrial Complex I Overcomes Chemoresistance in High OXPHOS Pancreatic Cancer. Cell Rep Med 2020; 1:100143. [PMID: 33294863 PMCID: PMC7691450 DOI: 10.1016/j.xcrm.2020.100143] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 08/28/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023]
Abstract
Mitochondrial respiration (oxidative phosphorylation, OXPHOS) is an emerging target in currently refractory cancers such as pancreatic ductal adenocarcinoma (PDAC). However, the variability of energetic metabolic adaptations between PDAC patients has not been assessed in functional investigations. In this work, we demonstrate that OXPHOS rates are highly heterogeneous between patient tumors, and that high OXPHOS tumors are enriched in mitochondrial respiratory complex I at protein and mRNA levels. Therefore, we treated PDAC cells with phenformin (complex I inhibitor) in combination with standard chemotherapy (gemcitabine), showing that this treatment is synergistic specifically in high OXPHOS cells. Furthermore, phenformin cooperates with gemcitabine in high OXPHOS tumors in two orthotopic mouse models (xenografts and syngeneic allografts). In conclusion, this work proposes a strategy to identify PDAC patients likely to respond to the targeting of mitochondrial energetic metabolism in combination with chemotherapy, and that phenformin should be clinically tested in appropriate PDAC patient subpopulations.
Collapse
Affiliation(s)
- Rawand Masoud
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Gabriela Reyes-Castellanos
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Sophie Lac
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Julie Garcia
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Samir Dou
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Laetitia Shintu
- Aix Marseille Université, CNRS, Centrale Marseille, ISM2, F-13013 Marseille, France
| | - Nadine Abdel Hadi
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Tristan Gicquel
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Abdessamad El Kaoutari
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Binta Diémé
- Aix Marseille Université, CNRS, Centrale Marseille, ISM2, F-13013 Marseille, France
| | - Fabrice Tranchida
- Aix Marseille Université, CNRS, Centrale Marseille, ISM2, F-13013 Marseille, France
| | - Laurie Cormareche
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Laurence Borge
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Odile Gayet
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Eddy Pasquier
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Nelson Dusetti
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Juan Iovanna
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Alice Carrier
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| |
Collapse
|
158
|
Chang HW, Lee M, Lee YS, Kim SH, Lee JC, Park JJ, Nam HY, Kim MR, Han MW, Kim SW, Kim SY. p53-dependent glutamine usage determines susceptibility to oxidative stress in radioresistant head and neck cancer cells. Cell Signal 2020; 77:109820. [PMID: 33137455 DOI: 10.1016/j.cellsig.2020.109820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 01/08/2023]
Abstract
The manner in which p53 maintains redox homeostasis and the means by which two key metabolic elements, glucose and glutamine, contribute to p53-dependent redox stability remain unclear. To elucidate the manner in which p53 deals with glucose-deprived, reactive oxygen species (ROS)-prone conditions in this regard, two isogenic cancer subclones (HN3R-A and HN3R-B) bearing distinct p53 mutations as an in vitro model of intratumoral p53 heterogeneity were identified. Following cumulative irradiation, the subclones showed a similar metabolic shift to aerobic glycolysis and increasing NADPH biogenesis for cellular defense against oxidative damage irrespective of p53 status. The radioresistant cancer cells became more sensitive to glycolysis-targeting drugs. However, in glucose-deprived and ROS-prone conditions, HN3R-B, the subclone with the original p53 increased the utilization of glutamine by GLS2, thereby maintaining redox homeostasis and ATP. Conversely, HN3R-A, the p53-deficient radioresistant subclone displayed an impairment in glutamine usage and high susceptibility to metabolic stresses as well as ROS-inducing agents despite the increased ROS scavenging system. Collectively, our findings suggest that p53 governs the alternative utilization of metabolic ingredients, such as glucose and glutamine, in ROS-prone conditions. Thus, p53 status may be an important biomarker for selecting cancer treatment strategies, including metabolic drugs and ROS-inducing agents, for recurrent cancers after radiotherapy.
Collapse
Affiliation(s)
- Hyo Won Chang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - MyungJin Lee
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoon Sun Lee
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Song Hee Kim
- Department of Otolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Jong Cheol Lee
- Department of Otolaryngology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, South Korea
| | - Jung Je Park
- Department of Otolaryngology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Hae Yun Nam
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi Ra Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Inje University College of Medicine, Haeundae Paik Hospital, Busan, Republic of Korea
| | - Myung Woul Han
- Department of Otolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Sang Yoon Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
159
|
Shi T, Dansen TB. Reactive Oxygen Species Induced p53 Activation: DNA Damage, Redox Signaling, or Both? Antioxid Redox Signal 2020; 33:839-859. [PMID: 32151151 DOI: 10.1089/ars.2020.8074] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Significance: The p53 tumor suppressor has been dubbed the "guardian of genome" because of its various roles in the response to DNA damage such as DNA damage repair, cell cycle arrest, senescence, and apoptosis, all of which are in place to prevent mutations from being passed on down the lineage. Recent Advances: Reactive oxygen species (ROS), for instance hydrogen peroxide derived from mitochondrial respiration, have long been regarded mainly as a major source of cellular damage to DNA and other macromolecules. Critical Issues: More recently, ROS have been shown to also play important physiological roles as second messengers in so-called redox signaling. It is, therefore, not clear whether the observed activation of p53 by ROS is mediated through the DNA damage response, redox signaling, or both. In this review, we will discuss the similarities and differences between p53 activation in response to DNA damage and redox signaling in terms of upstream signaling and downstream transcriptional program activation. Future Directions: Understanding whether and how DNA damage and redox signaling-dependent p53 activation can be dissected could be useful to develop anti-cancer therapeutic p53-reactivation strategies that do not depend on the induction of DNA damage and the resulting additional mutational load.
Collapse
Affiliation(s)
- Tao Shi
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tobias B Dansen
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
160
|
Digging deeper through glucose metabolism and its regulators in cancer and metastasis. Life Sci 2020; 264:118603. [PMID: 33091446 DOI: 10.1016/j.lfs.2020.118603] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022]
Abstract
Glucose metabolism enzymes and transporters play major role in cancer development and metastasis. In this study, we discuss glucose metabolism, transporters, receptors, hormones, oncogenes and tumor suppressors which interact with glucose metabolism and we try to discuss their major role in cancer development and cancer metabolism. We try to highlight the. Metabolic changes in cancer and metastasis upregulation of glycolysis is observed in many primary and metastatic cancers and aerobic glycolysis is the most favorable mechanism for glucose metabolism in cancer cells, and it is a kind of evolutionary change. The question that is posed at this juncture is: Can we use aerobic glycolysis phenotype and enzymes beyond this mechanism in estimating cancer prognosis and metastasis? Lactate is a metabolite of glucose metabolism and it is a key player in cancer and metastasis in both normoxic and hypoxic condition so lactate dehydrogenase can be a good prognostic biomarker. Furthermore, monocarboxylic transporter which is the main lactate transporter can be good target in therapeutic studies. Glycolysis enzymes are valuable enzymes in cancer and metastasis diagnosis and can be used as therapeutic targets in cancer treatment. Designing a diagnostic and prognostic profile for cancer metastasis seems to be possible base on glycolysis enzymes and glucose transporters. Also, glucose metabolism enzymes and agents can give us a clear vision in estimating cancer metastasis. We can promote a panel of genes that detect genetic changes in glucose metabolism agents to diagnose cancer metastasis.
Collapse
|
161
|
Broekman KE, Hof MAJ, Touw DJ, Gietema JA, Nijman HW, Lefrandt JD, Reyners AKL, Jalving M. Phase I study of metformin in combination with carboplatin/paclitaxel chemotherapy in patients with advanced epithelial ovarian cancer. Invest New Drugs 2020; 38:1454-1462. [PMID: 32146550 PMCID: PMC7497683 DOI: 10.1007/s10637-020-00920-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
Background Metformin use is associated with reduced cancer risk in epidemiological studies and has preclinical anti-cancer activity in ovarian cancer models. The primary objective of this phase I study was to determine the recommended phase II dose (RP2D) of metformin in combination with carboplatin/paclitaxel in patients with ovarian cancer. Secondary objectives were to describe safety and pharmacokinetics. Methods In this single-center trial the RP2D of metformin in combination with carboplatin area under the concentration-time curve (AUC) 6 and paclitaxel 175 mg/m2 every 3 weeks (q3w) in patients with advanced epithelial ovarian cancer was determined using a 3 + 3 escalation rule at three fixed dose levels: 500 mg three times daily (tds), 850 mg tds and 1000 mg tds. Metformin was commenced on day 3 of cycle 1 and continued until 3 weeks after the last chemotherapy administration. The RP2D was defined as the dose level at which 0 of 3 or ≤ 1 of 6 evaluable subjects experienced a metformin-related dose-limiting toxicity (DLT). Safety was assessed according to CTCAE v4.0. Plasma and serum samples for pharmacokinetic (PK) analyses were collected during treatment cycles 1 and 2. Results Fifteen patients with epithelial ovarian cancer and an indication for neo-adjuvant (n = 5) or palliative (n = 10) treatment were included. No DLTs were observed. Three patients discontinued study treatment during cycle 1 for other reasons than DLT. Six patients were treated at the RP2D of metformin 1000 mg tds. The most frequent low-grade toxicities were anemia, hypomagnesemia and diarrhea. Grade 3 adverse events (AEs) occurred in ten patients, most common were leucopenia (n = 4), thrombocytopenia (n = 3) and increased GGT (n = 3). There were no grade 4 AEs. Metformin increased the platinum (Pt) AUC (Δ22%, p = 0.013) and decreased the Pt clearance (Δ-28%, p = 0.013). Metformin plasma levels were all within the therapeutic range for diabetic patients (0.1-4 mg/L). Conclusion The RP2D of metformin in combination with carboplatin and paclitaxel in advanced ovarian cancer is 1000 mg tds. This is higher than the RP2D reported for combination with targeted agents. A potential PK interaction of metformin with carboplatin was identified.
Collapse
Affiliation(s)
- K Esther Broekman
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, the Netherlands
| | - Marieke A J Hof
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, the Netherlands
| | - Hans W Nijman
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joop D Lefrandt
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - An K L Reyners
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, the Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, the Netherlands.
| |
Collapse
|
162
|
Natural Agents Targeting Mitochondria in Cancer. Int J Mol Sci 2020; 21:ijms21196992. [PMID: 32977472 PMCID: PMC7582837 DOI: 10.3390/ijms21196992] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are the key energy provider to highly proliferating cancer cells, and are subsequently considered one of the critical targets in cancer therapeutics. Several compounds have been studied for their mitochondria-targeting ability in cancer cells. These studies’ outcomes have led to the invention of “mitocans”, a category of drug known to precisely target the cancer cells’ mitochondria. Based upon their mode of action, mitocans have been divided into eight classes. To date, different synthetic compounds have been suggested to be potential mitocans, but unfortunately, they are observed to exert adverse effects. Many studies have been published justifying the medicinal significance of large numbers of natural agents for their mitochondria-targeting ability and anticancer activities with minimal or no side effects. However, these natural agents have never been critically analyzed for their mitochondria-targeting activity. This review aims to evaluate the various natural agents affecting mitochondria and categorize them in different classes. Henceforth, our study may further support the potential mitocan behavior of various natural agents and highlight their significance in formulating novel potential anticancer therapeutics.
Collapse
|
163
|
Jung JH, Lee H, Zeng SX, Lu H. RBM10, a New Regulator of p53. Cells 2020; 9:cells9092107. [PMID: 32947864 PMCID: PMC7563659 DOI: 10.3390/cells9092107] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/14/2022] Open
Abstract
The tumor suppressor p53 acts as a transcription factor that regulates the expression of a number of genes responsible for DNA repair, cell cycle arrest, metabolism, cell migration, angiogenesis, ferroptosis, senescence, and apoptosis. It is the most commonly silenced or mutated gene in cancer, as approximately 50% of all types of human cancers harbor TP53 mutations. Activation of p53 is detrimental to normal cells, thus it is tightly regulated via multiple mechanisms. One of the recently identified regulators of p53 is RNA-binding motif protein 10 (RBM10). RBM10 is an RNA-binding protein frequently deleted or mutated in cancer cells. Its loss of function results in various deformities, such as cleft palate and malformation of the heart, and diseases such as lung adenocarcinoma. In addition, RBM10 mutations are frequently observed in lung adenocarcinomas, colorectal carcinomas, and pancreatic ductal adenocarcinomas. RBM10 plays a regulatory role in alternative splicing. Several recent studies not only linked this splicing regulation of RBM10 to cancer development, but also bridged RBM10's anticancer function to the p53 pathway. This review will focus on the current progress in our understanding of RBM10 regulation of p53, and its role in p53-dependent cancer prevention.
Collapse
MESH Headings
- Adenocarcinoma of Lung/genetics
- Adenocarcinoma of Lung/metabolism
- Adenocarcinoma of Lung/pathology
- Alternative Splicing
- Apoptosis/genetics
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Cycle Checkpoints/genetics
- Cell Movement
- Cell Proliferation
- Cellular Senescence
- Cleft Palate/genetics
- Cleft Palate/metabolism
- Cleft Palate/pathology
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Gene Expression Regulation, Neoplastic
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/metabolism
- Heart Defects, Congenital/pathology
- Humans
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Signal Transduction
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Ji Hoon Jung
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: or (J.H.J.); (H.L.); Tel.: +82-10-961-9597 (J.H.J.); +1-504-988-5293 (H.L.)
| | - Hyemin Lee
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (H.L.); (S.X.Z.)
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Shelya X Zeng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (H.L.); (S.X.Z.)
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hua Lu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (H.L.); (S.X.Z.)
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Correspondence: or (J.H.J.); (H.L.); Tel.: +82-10-961-9597 (J.H.J.); +1-504-988-5293 (H.L.)
| |
Collapse
|
164
|
Barcherini V, Almeida J, Lopes EA, Wang M, Magalhães E Silva D, Mori M, Wang S, Saraiva L, Santos MMM. Potency and Selectivity Optimization of Tryptophanol-Derived Oxazoloisoindolinones: Novel p53 Activators in Human Colorectal Cancer. ChemMedChem 2020; 16:250-258. [PMID: 32737944 DOI: 10.1002/cmdc.202000522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Indexed: 12/16/2022]
Abstract
To search for novel p53 activators, four series of novel (S)- and (R)-tryptophanol-derived oxazoloisoindolinones were synthesized in a straightforward manner and their antiproliferative activity was evaluated in the human colorectal cancer HCT116 cell line. Structural optimization of the hit compound SLMP53-1 led to the identification of a (R)-tryptophanol-derived isoindolinone that was found to be six-fold more active, with increased selectivity for HCT116 cells with p53 and with low toxicity in normal cells. Binding studies with MDM2 showed that the antiproliferative activity of tryptophanol-derived isoindolinones does not involve inhibition of the main negative regulator of the p53 protein. Molecular docking simulations showed that although these molecules establish hydrophobic interactions with MDM2, they do not possess the required features to bind MDM2.
Collapse
Affiliation(s)
- Valentina Barcherini
- Department of Therapeutic and Pharmaceutical Chemistry, University of Lisbon, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Joana Almeida
- Department of Biological Sciences, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Elizabeth A Lopes
- Department of Therapeutic and Pharmaceutical Chemistry, University of Lisbon, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Mi Wang
- Rogel Cancer Center, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - Diogo Magalhães E Silva
- Department of Therapeutic and Pharmaceutical Chemistry, University of Lisbon, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Shaomeng Wang
- Rogel Cancer Center, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lucília Saraiva
- Department of Biological Sciences, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Maria M M Santos
- Department of Therapeutic and Pharmaceutical Chemistry, University of Lisbon, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| |
Collapse
|
165
|
Tonoi T, Ikeda M, Sato T, Inohana T, Kawahara R, Murata T, Shiina I. Total Synthesis of the Antitumor Depsipeptide FE399 and Its S‐Benzyl Derivative: A Macrolactamization Approach. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Takayuki Tonoi
- Department of Applied Chemistry Faculty of Science Tokyo University of Science 1‐3 Kagurazaka, Shinjuku‐ku 162‐8601 Tokyo Japan
| | - Miyuki Ikeda
- Department of Applied Chemistry Faculty of Science Tokyo University of Science 1‐3 Kagurazaka, Shinjuku‐ku 162‐8601 Tokyo Japan
| | - Teruyuki Sato
- Department of Applied Chemistry Faculty of Science Tokyo University of Science 1‐3 Kagurazaka, Shinjuku‐ku 162‐8601 Tokyo Japan
| | - Takehiko Inohana
- Department of Applied Chemistry Faculty of Science Tokyo University of Science 1‐3 Kagurazaka, Shinjuku‐ku 162‐8601 Tokyo Japan
| | - Ryo Kawahara
- Department of Applied Chemistry Faculty of Science Tokyo University of Science 1‐3 Kagurazaka, Shinjuku‐ku 162‐8601 Tokyo Japan
| | - Takatsugu Murata
- Department of Applied Chemistry Faculty of Science Tokyo University of Science 1‐3 Kagurazaka, Shinjuku‐ku 162‐8601 Tokyo Japan
| | - Isamu Shiina
- Department of Applied Chemistry Faculty of Science Tokyo University of Science 1‐3 Kagurazaka, Shinjuku‐ku 162‐8601 Tokyo Japan
| |
Collapse
|
166
|
Kwon CY, Cho IH, Park KS. Therapeutic Effects and Mechanisms of Herbal Medicines for Treating Polycystic Ovary Syndrome: A Review. Front Pharmacol 2020; 11:1192. [PMID: 32903374 PMCID: PMC7434855 DOI: 10.3389/fphar.2020.01192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/22/2020] [Indexed: 12/19/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is one of the most common disorders of endocrinology in reproductive-age women. In this study, we reviewed data on the effects and underlying mechanisms of herbal medicines used in the treatment of PCOS in laboratory studies. Methods Articles published in English up to June 30, 2018 were searched in Medline and EMBASE. We extracted data regarding herbal intervention; target cell (or animal model) usage; method of herbal extraction; route of administration; dosage and periods; and outcomes of the compounds isolated from herbs, individual herbal extracts, and herbal formula decoctions. We summarized the actions and the mechanisms underlying the beneficial effects of herbal medicines on PCOS. Results A total of 27 studies involving 22 herbal medicines reported their efficacy on PCOS. The herbal interventions in the 27 studies comprised four compounds isolated from herbs (6 studies), nine individual herbal extracts (11 studies), and nine herbal formula decoctions (10 studies). Herbal medicines normalized female hormones, diminished male hormones, recovered the estrous cycle, ameliorated insulin resistance, and improved lipid metabolism in PCOS. The mechanisms underlying the beneficial effects of herbal medicines on PCOS were found to be associated with anti-inflammation, anti-oxidative stress, inhibition of autophagy and/or apoptosis, and ovarian nerve growth factor reduction. Conclusions Herbal medicines are thought to be promising resources in the development of effective therapeutic agents for PCOS. Further studies that include methodological quality assessment and quantitative synthesis of outcomes are recommended.
Collapse
Affiliation(s)
- Chan-Young Kwon
- Department of Oriental Neuropsychiatry, Dong-eui University College of Korean Medicine, Busan, South Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, Brain Korea 21 Plus Program, and Institute of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Kyoung Sun Park
- Jaseng Hospital of Korean Medicine, Seoul, South Korea.,Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, South Korea
| |
Collapse
|
167
|
Alam E, Maaliki L, Nasr Z. Ribosomal protein S3 selectively affects colon cancer growth by modulating the levels of p53 and lactate dehydrogenase. Mol Biol Rep 2020; 47:6083-6090. [PMID: 32748020 DOI: 10.1007/s11033-020-05683-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 07/26/2020] [Indexed: 01/10/2023]
Abstract
Ribosomal protein S3 (RPS3) is a component of the 40S ribosomal subunit. It is known to function in ribosome biogenesis and as an endonuclease. RPS3 has been shown to be over expressed in colon adenocarcinoma but its role in colon cancer is still unknown. In this study, we aim at determining the expression levels of RPS3 in a colon cancer cell line Caco-2 compared to a normal colon mucosa cell line NCM-460 and study the effects of targeting this protein by siRNA on cellular behavior. RPS3 was found to be expressed in both cell lines. However, siRNA treatment showed a more protruding effect on Caco-2 cells compared to NCM-460 cells. RPS3 knockdown led to a significant decrease in the proliferation, survival, migration and invasion and an increase in the apoptosis of Caco-2 cells. Western blot analysis demonstrated that these effects correlated with an increase in the level of the tumor suppressor p53 and a decrease in the level and activity of lactate dehydrogenase (LDH), an enzyme involved in the metabolism of cancer cells. No significant effect was shown in normal colon NCM-460 cells. Targeting p53 by siRNA did not affect RPS3 levels indicating that p53 may be a downstream target of RPS3. However, the concurrent knockdown of RPS3 and p53 showed no change in LDH level in Caco-2 cells suggesting an interesting interplay among the three proteins. These findings might present RPS3 as a selective molecular marker in colon cancer and an attractive target for colon cancer therapy.
Collapse
Affiliation(s)
- Elie Alam
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, P.O.B. 100, Tripoli, Lebanon
| | - Lama Maaliki
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, P.O.B. 100, Tripoli, Lebanon
| | - Zeina Nasr
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, P.O.B. 100, Tripoli, Lebanon.
| |
Collapse
|
168
|
Reyes-Castellanos G, Masoud R, Carrier A. Mitochondrial Metabolism in PDAC: From Better Knowledge to New Targeting Strategies. Biomedicines 2020; 8:biomedicines8080270. [PMID: 32756381 PMCID: PMC7460249 DOI: 10.3390/biomedicines8080270] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer cells reprogram their metabolism to meet bioenergetics and biosynthetic demands. The first observation of metabolic reprogramming in cancer cells was made a century ago (“Warburg effect” or aerobic glycolysis), leading to the classical view that cancer metabolism relies on a glycolytic phenotype. There is now accumulating evidence that most cancers also rely on mitochondria to satisfy their metabolic needs. Indeed, the current view of cancer metabolism places mitochondria as key actors in all facets of cancer progression. Importantly, mitochondrial metabolism has become a very promising target in cancer therapy, including for refractory cancers such as Pancreatic Ductal AdenoCarcinoma (PDAC). In particular, mitochondrial oxidative phosphorylation (OXPHOS) is an important target in cancer therapy. Other therapeutic strategies include the targeting of glutamine and fatty acids metabolism, as well as the inhibition of the TriCarboxylic Acid (TCA) cycle intermediates. A better knowledge of how pancreatic cancer cells regulate mitochondrial metabolism will allow the identification of metabolic vulnerabilities and thus novel and more efficient therapeutic options for the benefit of each patient.
Collapse
Affiliation(s)
| | | | - Alice Carrier
- Correspondence: ; Tel.: +33-491828829; Fax: +33-491826083
| |
Collapse
|
169
|
Yu H, Li M, He R, Fang P, Wang Q, Yi Y, Wang F, Zhou L, Zhang Y, Chen A, Peng N, Liu D, Trilling M, Broering R, Wiemer EAC, Lu M, Zhu Y, Liu S. Major Vault Protein Promotes Hepatocellular Carcinoma Through Targeting Interferon Regulatory Factor 2 and Decreasing p53 Activity. Hepatology 2020; 72:518-534. [PMID: 31758709 DOI: 10.1002/hep.31045] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/08/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND AIMS Major vault protein (MVP) is up-regulated during infections with hepatitis B virus (HBV) and hepatitis C virus (HCV). Here, we found that MVP deficiency inhibited hepatocellular carcinoma (HCC) development induced by diethylnitrosamine, hepatitis B X protein, and HCV core. APPROACH AND RESULTS Forced MVP expression was sufficient to induce HCC in mice. Mechanistic studies demonstrate that the ubiquitin ligase human double minute 2 (HDM2) forms mutual exclusive complexes either with interferon regulatory factor 2 (IRF2) or with p53. In the presence of MVP, HDM2 is liberated from IRF2, leading to the ubiquitination of the tumor suppressor p53. Mouse xenograft models showed that HBV and HCV promote carcinogenesis through MVP induction, resulting in a loss of p53 mediated by HDM2. Analyses of clinical samples from chronic hepatitis B, liver cirrhosis, and HCC revealed that MVP up-regulation correlates with several hallmarks of malignancy and associates with poor overall survival. CONCLUSIONS Taken together, through the sequestration of IRF2, MVP promotes an HDM2-dependent loss of p53 that promotes HCC development.
Collapse
Affiliation(s)
- Haisheng Yu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Mengqi Li
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Rui He
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Peining Fang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qiming Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Yu Yi
- The Key Laboratory of Biosystems Homeostasis and Protection of the Ministry of Education and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Li Zhou
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, School of Medicine, Wuhan University, Wuhan, China
| | - Yi Zhang
- Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Food and Pharmaceutical Engineering, Hubei University of Technology, Wuhan, China
| | - Aidong Chen
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Nanfang Peng
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Dan Liu
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ruth Broering
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Erik A C Wiemer
- Department of Medical Oncology, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
170
|
Dynamic regulation of histone H3 lysine (K) acetylation and deacetylation during prolonged oxygen deprivation in a champion anaerobe. Mol Cell Biochem 2020; 474:229-241. [PMID: 32729004 DOI: 10.1007/s11010-020-03848-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022]
Abstract
Trachemys scripta elegans can survive up to three months of absolute anoxia at 3 °C and recover with minimal cellular damage. Red-eared sliders employ various physiological and biochemical adaptations to survive anoxia with metabolic rate depression (MRD) being the most prominent adaptation. MRD is mediated by epigenetic, transcriptional, post-transcriptional, and post-translational mechanisms aimed at shutting down cellular processes that are not needed for anoxia survival, while reprioritizing ATP towards cell processes that are vital for anaerobiosis. Histone acetylation/deacetylation are epigenetic modifications that maintain a proper balance between permissive chromatin and restricted chromatin, yet very little is known about protein regulation and enzymatic activity of the writers and erasers of acetylation during natural anoxia tolerance. As such, this study explored the interplay between transcriptional activators, histone acetyltransferases (HATs), and transcriptional repressors, sirtuins (SIRTs), along with three prominent acetyl-lysine (K) moieties of histone H3 in the liver of red-eared sliders. Western immunoblotting was used to measure acetylation levels of H3-K14, H3-K18, and H3-K56, as well as protein levels of histone H3-total, HATs, and nuclear SIRTs in the liver in response to 5 h and 20 h anoxia. Global and nuclear enzymatic activity of HATs and enzymatic activity of nuclear SIRTs were also measured. Overall, a strong suppression of HATs-mediated H3 acetylation and SIRT-mediated deacetylation was evident in the liver of red-eared sliders that could play an important role in ATP conservation as part of the overall reduction in metabolic rate.
Collapse
|
171
|
Rodríguez C, Puente-Moncada N, Reiter RJ, Sánchez-Sánchez AM, Herrera F, Rodríguez-Blanco J, Duarte-Olivenza C, Turos-Cabal M, Antolín I, Martín V. Regulation of cancer cell glucose metabolism is determinant for cancer cell fate after melatonin administration. J Cell Physiol 2020; 236:27-40. [PMID: 32725819 DOI: 10.1002/jcp.29886] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/02/2020] [Accepted: 06/06/2020] [Indexed: 12/30/2022]
Abstract
Several oncogenic pathways plus local microenvironmental conditions, such as hypoxia, converge on the regulation of cancer cells metabolism. The major metabolic alteration consists of a shift from oxidative phosphorylation as the major glucose consumer to aerobic glycolysis, although most of cancer cells utilize both pathways to a greater or lesser extent. Aerobic glycolysis, together with the directly related metabolic pathways such as the tricarboxylic acid cycle, the pentose phosphate pathway, or gluconeogenesis are currently considered as therapeutic targets in cancer research. Melatonin has been reported to present numerous antitumor effects, which result in a reduced cell growth. This is achieved with both low and high concentrations with no relevant side effects. Indeed, high concentrations of this indolamine reduce proliferation of cancer types resistant to low concentrations and induce cell death in some types of tumors. Previous work suggest that regulation of glucose metabolism and other related pathways play an important role in the antitumoral effects of high concentration of melatonin. In the present review, we analyze recent work on the regulation by such concentrations of this indolamine on aerobic glycolysis, gluconeogenesis, the tricarboxylic acid cycle and the pentose phosphate pathways of cancer cells.
Collapse
Affiliation(s)
- Carmen Rodríguez
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,University Institute of Oncology of the Principality of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of the Principality of Asturias (ISPA), University of Oviedo, Oviedo, Spain
| | - Noelia Puente-Moncada
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,University Institute of Oncology of the Principality of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of the Principality of Asturias (ISPA), University of Oviedo, Oviedo, Spain
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas
| | - Ana M Sánchez-Sánchez
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,University Institute of Oncology of the Principality of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of the Principality of Asturias (ISPA), University of Oviedo, Oviedo, Spain
| | - Federico Herrera
- Cell Structure and Dynamics Laboratory, Institute of Chemical and Biological Technology (ITQB-NOVA), Estação Agronómica Nacional, Oeiras, Portugal
| | - Jezabel Rodríguez-Blanco
- Molecular Oncology Program, Department of Surgery, The DeWitt Daughtry Family, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Cristina Duarte-Olivenza
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,University Institute of Oncology of the Principality of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of the Principality of Asturias (ISPA), University of Oviedo, Oviedo, Spain
| | - María Turos-Cabal
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,University Institute of Oncology of the Principality of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of the Principality of Asturias (ISPA), University of Oviedo, Oviedo, Spain
| | - Isaac Antolín
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,University Institute of Oncology of the Principality of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of the Principality of Asturias (ISPA), University of Oviedo, Oviedo, Spain
| | - Vanesa Martín
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,University Institute of Oncology of the Principality of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of the Principality of Asturias (ISPA), University of Oviedo, Oviedo, Spain
| |
Collapse
|
172
|
Patel KR, Patel HD. p53: An Attractive Therapeutic Target for Cancer. Curr Med Chem 2020; 27:3706-3734. [PMID: 31223076 DOI: 10.2174/1573406415666190621094704] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/28/2019] [Accepted: 04/16/2019] [Indexed: 02/08/2023]
Abstract
Cancer is a leading cause of death worldwide. It initiates when cell cycle regulatory genes lose their function either by environmental and/or by internal factors. Tumor suppressor protein p53, known as "Guardian of genome", plays a central role in maintaining genomic stability of the cell. Mutation of TP53 is documented in more than 50% of human cancers, usually by overexpression of negative regulator protein MDM2. Hence, reactivation of p53 by blocking the protein-protein interaction between the murine double minute 2 (MDM2) and the tumor suppressor protein p53 has become the most promising therapeutic strategy in oncology. Several classes of small molecules have been identified as potent, selective and efficient p53-MDM2 inhibitors. Herein, we review the druggability of p53-MDM2 inhibitors and their optimization approaches as well as clinical candidates categorized by scaffold type.
Collapse
Affiliation(s)
- Krupa R Patel
- Department of Chemistry, School of Sciences, Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Hitesh D Patel
- Department of Chemistry, School of Sciences, Gujarat University, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
173
|
Wang H, Wang X, Xu L, Zhang J, Cao H. High expression levels of pyrimidine metabolic rate-limiting enzymes are adverse prognostic factors in lung adenocarcinoma: a study based on The Cancer Genome Atlas and Gene Expression Omnibus datasets. Purinergic Signal 2020; 16:347-366. [PMID: 32638267 PMCID: PMC7524999 DOI: 10.1007/s11302-020-09711-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Reprogramming of metabolism is described in many types of cancer and is associated with the clinical outcomes. However, the prognostic significance of pyrimidine metabolism signaling pathway in lung adenocarcinoma (LUAD) is unclear. Using the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) datasets, we found that the pyrimidine metabolism signaling pathway was significantly enriched in LUAD. Compared with normal lung tissues, the pyrimidine metabolic rate–limiting enzymes were highly expressed in lung tumor tissues. The high expression levels of pyrimidine metabolic–rate limiting enzymes were associated with unfavorable prognosis. However, purinergic receptors P2RX1, P2RX7, P2RY12, P2RY13, and P2RY14 were relatively downregulated in lung cancer tissues and were associated with favorable prognosis. Moreover, we found that hypo-DNA methylation, DNA amplification, and TP53 mutation were contributing to the high expression levels of pyrimidine metabolic rate–limiting enzymes in lung cancer cells. Furthermore, combined pyrimidine metabolic rate–limiting enzymes had significant prognostic effects in LUAD. Comprehensively, the pyrimidine metabolic rate–limiting enzymes were highly expressed in bladder cancer, breast cancer, colon cancer, liver cancer, and stomach cancer. And the high expression levels of pyrimidine metabolic rate–limiting enzymes were associated with unfavorable prognosis in liver cancer. Overall, our results suggested the mRNA levels of pyrimidine metabolic rate–limiting enzymes CAD, DTYMK, RRM1, RRM2, TK1, TYMS, UCK2, NR5C2, and TK2 were predictive of lung cancer as well as other cancers.
Collapse
Affiliation(s)
- Haiwei Wang
- Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Maternity and Child Health Hospital,, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.
- Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate,, National Health and Family Planning Commission, Fuzhou, Fujian, China.
| | - Xinrui Wang
- Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Maternity and Child Health Hospital,, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate,, National Health and Family Planning Commission, Fuzhou, Fujian, China
| | - Liangpu Xu
- Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Maternity and Child Health Hospital,, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate,, National Health and Family Planning Commission, Fuzhou, Fujian, China
| | - Ji Zhang
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital Affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Hua Cao
- Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Maternity and Child Health Hospital,, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.
- Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate,, National Health and Family Planning Commission, Fuzhou, Fujian, China.
| |
Collapse
|
174
|
Mumyatova VA, Balakina AA, Lapshina MA, Sen' VD, Kornev AB, Terent'ev AA. Influence of Tumor Suppressor p53 Functioning on the Expression of Antioxidant System Genes under the Action of Cytotoxic Compounds. Bull Exp Biol Med 2020; 169:169-175. [PMID: 32504383 DOI: 10.1007/s10517-020-04844-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Indexed: 01/18/2023]
Abstract
The effect of inhibition of the tumor suppressor p53 on the antioxidant system genes expression under the influence of cytotoxic compounds of the platinum group was studied. It was found that the action of platinum(II) and platinum(IV) complexes induced accumulation of p53 protein with a maximum in 12 h, which was confirmed by an increase in the expression of the P21 gene, the target gene of the p53 protein. It was shown that the action of platinum complexes activated the expression of catalase and superoxide dismutase 2 genes. Suppression of p53 protein functions with specific inhibitor α-piphitrin under the action of platinum complexes reduced the expression of catalase and superoxide dismutase 2 genes and the target gene P21, which attested to the p53-dependent regulation of these genes.
Collapse
Affiliation(s)
- V A Mumyatova
- Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Moscow region, Russia.
- Scientific and Educational Center in Chernogolovka of Moscow Region State University, Mytishchi, Moscow region, Russia.
| | - A A Balakina
- Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Moscow region, Russia
- Scientific and Educational Center in Chernogolovka of Moscow Region State University, Mytishchi, Moscow region, Russia
| | - M A Lapshina
- Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Moscow region, Russia
- Scientific and Educational Center in Chernogolovka of Moscow Region State University, Mytishchi, Moscow region, Russia
| | - V D Sen'
- Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Moscow region, Russia
| | - A B Kornev
- Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Moscow region, Russia
| | - A A Terent'ev
- Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Moscow region, Russia
- M. V. Lomonosov Moscow State University, Moscow, Russia
- Scientific and Educational Center in Chernogolovka of Moscow Region State University, Mytishchi, Moscow region, Russia
| |
Collapse
|
175
|
Chappell WH, Candido S, Abrams SL, Akula SM, Steelman LS, Martelli AM, Ratti S, Cocco L, Cervello M, Montalto G, Nicoletti F, Libra M, McCubrey JA. Influences of TP53 and the anti-aging DDR1 receptor in controlling Raf/MEK/ERK and PI3K/Akt expression and chemotherapeutic drug sensitivity in prostate cancer cell lines. Aging (Albany NY) 2020; 12:10194-10210. [PMID: 32492656 PMCID: PMC7346063 DOI: 10.18632/aging.103377] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/20/2020] [Indexed: 12/20/2022]
Abstract
Background: TP53 plays critical roles in sensitivity to chemotherapy, and aging. Collagen is very important in aging. The molecular structure and biochemical properties of collagen changes during aging. The discoidin domain receptor (DDR1) is regulated in part by collagen. Elucidating the links between TP53 and DDR1 in chemosensitivity and aging could improve therapies against cancer and aging. Results: Restoration of WT-TP53 activity resulted in increased sensitivity to chemotherapeutic drugs and elevated expression of key components of the Raf/MEK/ERK, PI3K/Akt and DDR1 pathways. DDR1 could modulate the levels of Raf/MEK/ERK and PI3K/Akt pathways as well as sensitize the cells to chemotherapeutic drugs. In contrast, suppression of WT TP53 with a dominant negative (DN) TP53 gene, suppressed DDR1 protein levels and increased their chemoresistance. Conclusion: Restoration of WT TP53 activity or increased expression of the anti-aging DDR1 collagen receptor can result in enhanced sensitivity to chemotherapeutic drugs. Our innovative studies indicate the important links between WT TP53 and DDR1 which can modulate Raf/MEK/ERK and PI3K/Akt signaling as well as chemosensitivity and aging. Methods: We investigated the roles of wild type (WT) and mutant TP53 on drug sensitivity of prostate cancer cells and the induction of Raf/MEK/ERK, PI3K/Akt and DDR1 expression and chemosensitivity.
Collapse
Affiliation(s)
- William H Chappell
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.,Current Address: Becton, Dickinson and Company (BD), BD Diagnostics, Franklin Lakes, NJ 07417, USA
| | - Saverio Candido
- Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Shaw M Akula
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Giuseppe Montalto
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy.,Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Massimo Libra
- Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
176
|
Wilkie MD, Anaam EA, Lau AS, Rubbi CP, Jones TM, Boyd MT, Vlatković N. TP53 mutations in head and neck cancer cells determine the Warburg phenotypic switch creating metabolic vulnerabilities and therapeutic opportunities for stratified therapies. Cancer Lett 2020; 478:107-121. [PMID: 32113989 PMCID: PMC7133053 DOI: 10.1016/j.canlet.2020.02.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/08/2020] [Accepted: 02/24/2020] [Indexed: 12/30/2022]
Abstract
Patients with mutated TP53 have been identified as having comparatively poor outcomes compared to those retaining wild-type p53 in many cancers, including squamous cell carcinomas of the head and neck (SCCHN). We have examined the role of p53 in regulation of metabolism in SCCHN cells and find that loss of p53 function determines the Warburg effect in these cells. Moreover, this metabolic adaptation to loss of p53 function creates an Achilles' heel for tumour cells that can be exploited for potential therapeutic benefit. Specifically, cells lacking normal wild-type p53 function, whether through mutation or RNAi-mediated downregulation, display a lack of metabolic flexibility, becoming more dependent on glycolysis and losing the ability to increase energy production from oxidative phosphorylation. Thus, cells that have compromised p53 function can be sensitised to ionizing radiation by pre-treatment with a glycolytic inhibitor. These results demonstrate the deterministic role of p53 in regulating energy metabolism and provide proof of principle evidence for an opportunity for patient stratification based on p53 status that can be exploited therapeutically using current standard of care treatment with ionising radiation.
Collapse
Affiliation(s)
- Mark D Wilkie
- Department of Molecular & Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK; Department of Otorhinolaryngology - Head & Neck Surgery, University Hospital Aintree, Lower Lane, Liverpool, L9 7AL, UK
| | - Emad A Anaam
- Department of Molecular & Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK
| | - Andrew S Lau
- Department of Molecular & Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK; Department of Otorhinolaryngology - Head & Neck Surgery, University Hospital Aintree, Lower Lane, Liverpool, L9 7AL, UK
| | - Carlos P Rubbi
- Department of Molecular & Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK
| | - Terence M Jones
- Department of Molecular & Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK; Department of Otorhinolaryngology - Head & Neck Surgery, University Hospital Aintree, Lower Lane, Liverpool, L9 7AL, UK
| | - Mark T Boyd
- Department of Molecular & Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK
| | - Nikolina Vlatković
- Department of Molecular & Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK.
| |
Collapse
|
177
|
Wang H, Wang X, Xu L, Cao H. Identification of transcription factors MYC and C/EBPβ mediated regulatory networks in heart failure based on gene expression omnibus datasets. BMC Cardiovasc Disord 2020; 20:250. [PMID: 32460775 PMCID: PMC7251862 DOI: 10.1186/s12872-020-01527-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/13/2020] [Indexed: 11/29/2022] Open
Abstract
Background Heart failure is one of leading cause of death worldwide. However, the transcriptional profiling of heart failure is unclear. Moreover, the signaling pathways and transcription factors involving the heart failure development also are largely unknown. Using published Gene Expression Omnibus (GEO) datasets, in the present study, we aim to comprehensively analyze the differentially expressed genes in failing heart tissues, and identified the critical signaling pathways and transcription factors involving heart failure development. Methods The transcriptional profiling of heart failure was identified from previously published gene expression datasets deposited in GSE5406, GSE16499 and GSE68316. The enriched signaling pathways and transcription factors were analyzed using Database for Annotation, Visualization and Integrated Discovery (DAVID) website and gene set enrichment analysis (GSEA) assay. The transcriptional networks were created by Cytoscape. Results Compared with the normal heart tissues, 90 genes were particularly differentially expressed in failing heart tissues, and those genes were associated with multiple metabolism signaling pathways and insulin signaling pathway. Metabolism and insulin signaling pathway were both inactivated in failing heart tissues. Transcription factors MYC and C/EBPβ were both negatively associated with the expression profiling of failing heart tissues in GSEA assay. Moreover, compared with normal heart tissues, MYC and C/EBPβ were down regulated in failing heart tissues. Furthermore, MYC and C/EBPβ mediated downstream target genes were also decreased in failing heart tissues. MYC and C/EBPβ were positively correlated with each other. At last, we constructed MYC and C/EBPβ mediated regulatory networks in failing heart tissues, and identified the MYC and C/EBPβ target genes which had been reported involving the heart failure developmental progress. Conclusions Our results suggested that metabolism pathways and insulin signaling pathway, transcription factors MYC and C/EBPβ played critical roles in heart failure developmental progress.
Collapse
Affiliation(s)
- Haiwei Wang
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, FuZhou, 350001, FuJian, China
| | - Xinrui Wang
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, FuZhou, 350001, FuJian, China
| | - Liangpu Xu
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, FuZhou, 350001, FuJian, China
| | - Hua Cao
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, FuZhou, 350001, FuJian, China.
| |
Collapse
|
178
|
Lynch-Sutherland CF, Chatterjee A, Stockwell PA, Eccles MR, Macaulay EC. Reawakening the Developmental Origins of Cancer Through Transposable Elements. Front Oncol 2020; 10:468. [PMID: 32432029 PMCID: PMC7214541 DOI: 10.3389/fonc.2020.00468] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/16/2020] [Indexed: 12/12/2022] Open
Abstract
Transposable elements (TEs) have an established role as important regulators of early human development, functioning as tissue-specific genes and regulatory elements. Functional TEs are highly active during early development, and interact with important developmental genes, some of which also function as oncogenes. Dedifferentiation is a hallmark of cancer, and is characterized by genetic and epigenetic changes that enable proliferation, self-renewal and a metabolism reminiscent of embryonic stem cells. There is also compelling evidence suggesting that the path to dedifferentiation in cancer can contribute to invasion and metastasis. TEs are frequently expressed in cancer, and recent work has identified a newly proposed mechanism involving extensive recruitment of TE-derived promoters to drive expression of oncogenes and subsequently promote oncogenesis—a process termed onco-exaptation. However, the mechanism by which this phenomenon occurs, and the extent to which it contributes to oncogenesis remains unknown. Initial hypotheses have proposed that onco-exaptation events are cancer-specific and arise randomly due to the dysregulated and hypomethylated state of cancer cells and abundance of TEs across the genome. However, we suspect that exaptation-like events may not just arise due to chance activation of novel regulatory relationships as proposed previously, but as a result of the reestablishment of early developmental regulatory relationships. Dedifferentiation in cancer is well-documented, along with expression of TEs. The known interactions between TEs and pluripotency factors such as NANOG and OCTt4 during early development, along with the expression of some placental-specific TE-derived transcripts in cancer support a possible link between TEs and dedifferentiation of tumor cells. Thus, we hypothesize that onco-exaptation events can be associated with the epigenetic reawakening of early developmental TEs to regulate expression of oncogenes and promote oncogenesis. We also suspect that activation of these early developmental regulatory TEs may promote dedifferentiation, although at this stage it is hard to predict whether TE activation is one of the initial drivers of dedifferentiation. We expect that developmental TE activation occurs as a result of the establishment of an epigenetic landscape in cancer that resembles that of early development and that developmental TE activation may also enable cancers to exploit early developmental pathways, repurposing them to promote malignancy.
Collapse
Affiliation(s)
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Peter A Stockwell
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Michael R Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Erin C Macaulay
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| |
Collapse
|
179
|
Li W, Wang J. Uncovering the Underlying Mechanisms of Cancer Metabolism through the Landscapes and Probability Flux Quantifications. iScience 2020; 23:101002. [PMID: 32276228 PMCID: PMC7150521 DOI: 10.1016/j.isci.2020.101002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/03/2019] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer metabolism is critical for understanding the mechanism of tumorigenesis, yet the understanding is still challenging. We studied gene-metabolism regulatory interactions and quantified the global driving forces for cancer-metabolism dynamics as the underlying landscape and probability flux. We uncovered four steady-state attractors: a normal state attractor, a cancer OXPHOS state attractor, a cancer glycolysis state attractor, and an intermediate cancer state attractor. We identified the key regulatory interactions through global sensitivity analysis based on the landscape topography. Different landscape topographies of glycolysis switch between normal cells and cancer cells were identified. We uncovered that the normal state to cancer state transformation is associated with the peaks of the probability flux and the thermodynamic dissipation, giving dynamical and thermodynamic origin of cancer formation. We found that cancer metabolism oscillations consume more energy to support cancer malignancy. This study provides a quantitative understanding of cancer metabolism and suggests a metabolic therapeutic strategy.
Collapse
Affiliation(s)
- Wenbo Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Jin Wang
- Department of Chemistry and Physics, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA.
| |
Collapse
|
180
|
Inhibition of Kv10.1 Channels Sensitizes Mitochondria of Cancer Cells to Antimetabolic Agents. Cancers (Basel) 2020; 12:cancers12040920. [PMID: 32283712 PMCID: PMC7226288 DOI: 10.3390/cancers12040920] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Reprogramming of energy metabolism constitutes one of the hallmarks of cancer and is, therefore, an emerging therapeutic target. We describe here that the potassium channel Kv10.1, which is frequently overexpressed in primary and metastatic cancer, and has been proposed a therapeutic target, participates in metabolic adaptation of cancer cells through regulation of mitochondrial dynamics. We used biochemical and cell biological techniques, live cell imaging and high-resolution microscopy, among other approaches, to study the impact of Kv10.1 on the regulation of mitochondrial stability. Inhibition of Kv10.1 expression or function led to mitochondrial fragmentation, increase in reactive oxygen species and increased autophagy. Cells with endogenous overexpression of Kv10.1 were also more sensitive to mitochondrial metabolism inhibitors than cells with low expression, indicating that they are more dependent on mitochondrial function. Consistently, a combined therapy using functional monoclonal antibodies for Kv10.1 and mitochondrial metabolism inhibitors resulted in enhanced efficacy of the inhibitors. Our data reveal a new mechanism regulated by Kv10.1 in cancer and a novel strategy to overcome drug resistance in cancers with a high expression of Kv10.1.
Collapse
|
181
|
Deng L, Yao P, Li L, Ji F, Zhao S, Xu C, Lan X, Jiang P. p53-mediated control of aspartate-asparagine homeostasis dictates LKB1 activity and modulates cell survival. Nat Commun 2020; 11:1755. [PMID: 32273511 PMCID: PMC7145870 DOI: 10.1038/s41467-020-15573-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 03/17/2020] [Indexed: 12/31/2022] Open
Abstract
Asparagine synthetase (ASNS) catalyses the ATP-dependent conversion of aspartate to asparagine. However, both the regulation and biological functions of asparagine in tumour cells remain largely unknown. Here, we report that p53 suppresses asparagine synthesis through the transcriptional downregulation of ASNS expression and disrupts asparagine-aspartate homeostasis, leading to lymphoma and colon tumour growth inhibition in vivo and in vitro. Moreover, the removal of asparagine from culture medium or the inhibition of ASNS impairs cell proliferation and induces p53/p21-dependent senescence and cell cycle arrest. Mechanistically, asparagine and aspartate regulate AMPK-mediated p53 activation by physically binding to LKB1 and oppositely modulating LKB1 activity. Thus, we found that p53 regulates asparagine metabolism and dictates cell survival by generating an auto-amplification loop via asparagine-aspartate-mediated LKB1-AMPK signalling. Our findings highlight a role for LKB1 in sensing asparagine and aspartate and connect asparagine metabolism to the cellular signalling transduction network that modulates cell survival.
Collapse
Affiliation(s)
- Longfei Deng
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.,School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Pengbo Yao
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.,School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Le Li
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.,School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Fansen Ji
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.,School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Shuang Zhao
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.,School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Chang Xu
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.,School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Xun Lan
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.,School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Peng Jiang
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China. .,School of Life Sciences, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
182
|
Coleman MF, Cozzo AJ, Pfeil AJ, Etigunta SK, Hursting SD. Cell Intrinsic and Systemic Metabolism in Tumor Immunity and Immunotherapy. Cancers (Basel) 2020; 12:cancers12040852. [PMID: 32244756 PMCID: PMC7225951 DOI: 10.3390/cancers12040852] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has shown extraordinary promise at treating cancers otherwise resistant to treatment. However, for ICI therapy to be effective, it must overcome the metabolic limitations of the tumor microenvironment. Tumor metabolism has long been understood to be highly dysregulated, with potent immunosuppressive effects. Moreover, T cell activation and longevity within the tumor microenvironment are intimately tied to T cell metabolism and are required for the long-term efficacy of ICI therapy. We discuss in this review the intersection of metabolic competition in the tumor microenvironment, T cell activation and metabolism, the roles of tumor cell metabolism in immune evasion, and the impact of host metabolism in determining immune surveillance and ICI therapy outcomes. We also discussed the effects of obesity and calorie restriction—two important systemic metabolic perturbations that impact intrinsic metabolic pathways in T cells as well as cancer cells.
Collapse
Affiliation(s)
- Michael F. Coleman
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
| | - Alyssa J. Cozzo
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
- Department of Medicine, Duke University, Durham, NC 27705, USA
| | - Alexander J. Pfeil
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
| | - Suhas K. Etigunta
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27516, USA
- Correspondence:
| |
Collapse
|
183
|
Jin C, Zhu X, Wu H, Wang Y, Hu X. Perturbation of phosphoglycerate kinase 1 (PGK1) only marginally affects glycolysis in cancer cells. J Biol Chem 2020; 295:6425-6446. [PMID: 32217690 DOI: 10.1074/jbc.ra119.012312] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
Phosphoglycerate kinase 1 (PGK1) plays important roles in glycolysis, yet its forward reaction kinetics are unknown, and its role especially in regulating cancer cell glycolysis is unclear. Here, we developed an enzyme assay to measure the kinetic parameters of the PGK1-catalyzed forward reaction. The Km values for 1,3-bisphosphoglyceric acid (1,3-BPG, the forward reaction substrate) were 4.36 μm (yeast PGK1) and 6.86 μm (human PKG1). The Km values for 3-phosphoglycerate (3-PG, the reverse reaction substrate and a serine precursor) were 146 μm (yeast PGK1) and 186 μm (human PGK1). The V max of the forward reaction was about 3.5- and 5.8-fold higher than that of the reverse reaction for the human and yeast enzymes, respectively. Consistently, the intracellular steady-state concentrations of 3-PG were between 180 and 550 μm in cancer cells, providing a basis for glycolysis to shuttle 3-PG to the serine synthesis pathway. Using siRNA-mediated PGK1-specific knockdown in five cancer cell lines derived from different tissues, along with titration of PGK1 in a cell-free glycolysis system, we found that the perturbation of PGK1 had no effect or only marginal effects on the glucose consumption and lactate generation. The PGK1 knockdown increased the concentrations of fructose 1,6-bisphosphate, dihydroxyacetone phosphate, glyceraldehyde 3-phosphate, and 1,3-BPG in nearly equal proportions, controlled by the kinetic and thermodynamic states of glycolysis. We conclude that perturbation of PGK1 in cancer cells insignificantly affects the conversion of glucose to lactate in glycolysis.
Collapse
Affiliation(s)
- Chengmeng Jin
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xiaobing Zhu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hao Wu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yuqi Wang
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xun Hu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| |
Collapse
|
184
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are two of the most common liver diseases associated with obesity, type 2 diabetes and metabolic syndrome. The prevalence of these conditions are increasingly rising and presently there is not a pharmacological option available in the market. Elucidation of the mechanism of action and the molecular underpinnings behind liver disease could help to better understand the pathophysiology of these illnesses. In this sense, in the last years modulation of the ghrelin system in preclinical animal models emerge as a promising therapeutic tool. In this review, we compile the latest knowledge of the modulation of ghrelin system and its intracellular pathways that regulates lipid metabolism, hepatic inflammation and liver fibrosis. We also describe novel processes implicated in the regulation of liver disease by ghrelin, such as autophagy or dysregulated circadian rhythms. In conclusion, the information displayed in this review support that the ghrelin system could be an appealing strategy for the treatment of liver disease.
Collapse
Affiliation(s)
- Mar Quiñones
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Omar Al-Massadi
- Inserm UMR-S1270, 75005, Paris, France.
- Faculté des Sciences et d'Ingénierie, Sorbonne Université, 75005, Paris, France.
- Institut du Fer a Moulin, Inserm, 17 rue du Fer à Moulin, 75005, Paris, France.
| |
Collapse
|
185
|
Phase I studies of vorinostat with ixazomib or pazopanib imply a role of antiangiogenesis-based therapy for TP53 mutant malignancies. Sci Rep 2020; 10:3080. [PMID: 32080210 PMCID: PMC7033174 DOI: 10.1038/s41598-020-58366-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
We performed two phase I trials of the histone deacetylase inhibitor vorinostat combined with either the vascular endothelial growth factor inhibitor pazopanib (NCT01339871) or the proteasome inhibitor ixazomib (NCT02042989) in patients with metastatic TP53 mutant solid tumors. Both trials followed a 3 + 3 dose-escalation design allowing for a dose expansion cohort of up to 14 additional patients with a specific tumor type. Patients had to have a confirmed TP53 mutation to be enrolled in NCT02042989. Among patients enrolled in NCT01339871, TP53 mutation status was determined for those for whom tumor specimens were available. The results of NCT01339871 were reported previously. Common treatment-related adverse events in NCT02042989 included anemia, thrombocytopenia, fatigue, nausea, vomiting, and diarrhea. Compared with patients with metastatic TP53 hotspot mutant solid tumors who were treated with ixazomib and vorinostat (n = 59), those who were treated with pazopanib and vorinostat (n = 11) had a significantly higher rate of clinical benefit, defined as stable disease lasting ≥6 months or an objective response (3.4% vs. 45%; p < 0.001), a significantly longer median progression-free survival duration (1.7 months [95% confidence interval (CI), 1.1–2.3] vs. 3.5 months [95% CI, 1.7–5.2]; p = 0.002), and a longer median overall survival duration (7.3 months [95% CI, 4.8–9.8] vs. 12.7 months [95% CI, 7.1–18.3]; p = 0.24). Our two phase I trials provide preliminary evidence supporting the use of antiangiogenisis-based therapy in patients with metastatic TP53 mutant solid tumors, especially in those with metastatic sarcoma or metastatic colorectal cancer.
Collapse
|
186
|
Castelli M, Piobbico D, Chiacchiaretta M, Brunacci C, Pieroni S, Bartoli D, Gargaro M, Fallarino F, Puccetti P, Soddu S, Della‐Fazia MA, Servillo G. HOPS/TMUB1 retains p53 in the cytoplasm and sustains p53-dependent mitochondrial apoptosis. EMBO Rep 2020; 21:e48073. [PMID: 31867855 PMCID: PMC7001502 DOI: 10.15252/embr.201948073] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 11/15/2019] [Accepted: 12/02/2019] [Indexed: 12/26/2022] Open
Abstract
Apoptotic signalling by p53 occurs at both transcriptional and non-transcriptional levels, as p53 may act as a direct apoptogenic stimulus via activation of the intrinsic mitochondrial pathway. HOPS is a highly conserved, ubiquitously expressed shuttling protein with an ubiquitin-like domain. We generated Hops-/- mice and observed that they are viable with no apparent phenotypic defects. However, when treated with chemotherapeutic agents, Hops-/- mice display a significant reduction in apoptosis, suggesting an impaired ability to respond to genotoxic stressors. We show that HOPS acts as a regulator of cytoplasmic p53 levels and function. By binding p53, HOPS inhibits p53 proteasomal degradation and favours p53 recruitment to mitochondria and apoptosis induction. By interfering with importin α, HOPS further increases p53 cytoplasmic levels. Thus, HOPS promotes the p53-dependent mitochondrial apoptosis pathway by preserving cytoplasmic p53 from both degradation and nuclear uptake.
Collapse
Affiliation(s)
- Marilena Castelli
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Danilo Piobbico
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | | | - Cinzia Brunacci
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Stefania Pieroni
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Daniela Bartoli
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Marco Gargaro
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Francesca Fallarino
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
- Centro Universitario di Ricerca sulla Genomica Funzionale (C.U.R.Ge.F.)University of PerugiaPerugiaItaly
| | - Paolo Puccetti
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
- Centro Universitario di Ricerca sulla Genomica Funzionale (C.U.R.Ge.F.)University of PerugiaPerugiaItaly
| | - Silvia Soddu
- Unit of Cellular Networks and Molecular Therapeutic TargetsIRCCS – Regina Elena National Cancer InstituteRomeItaly
| | | | - Giuseppe Servillo
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
- Centro Universitario di Ricerca sulla Genomica Funzionale (C.U.R.Ge.F.)University of PerugiaPerugiaItaly
| |
Collapse
|
187
|
Sarlak S, Lalou C, Amoedo ND, Rossignol R. Metabolic reprogramming by tobacco-specific nitrosamines (TSNAs) in cancer. Semin Cell Dev Biol 2020; 98:154-166. [PMID: 31699542 DOI: 10.1016/j.semcdb.2019.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 08/25/2019] [Accepted: 09/02/2019] [Indexed: 12/21/2022]
Abstract
Metabolic reprogramming is a hallmark of cancer and the link between oncogenes activation, tumor supressors inactivation and bioenergetics modulation is well established. However, numerous carcinogenic environmental factors are responsible for early cancer initiation and their impact on metabolic reprogramming just starts to be deciphered. For instance, it was recently shown that UVB irradiation triggers metabolic reprogramming at the pre-cancer stage with implication for skin cancer detection and therapy. These observations foster the need to study the early changes in tissue metabolism following exposure to other carcinogenic events. According to the International Agency for Research on Cancer (IARC), tobacco smoke is a major class I-carcinogenic environmental factor that contains different carcinogens, but little is known on the impact of tobacco smoke on tissue metabolism and its participation to cancer initiation. In particular, tobacco-specific nitrosamines (TSNAs) play a central role in tobacco-smoke mediated cancer initiation. Here we describe the recent advances that have led to a new hypothesis regarding the link between nitrosamines signaling and metabolic reprogramming in cancer.
Collapse
Affiliation(s)
- Saharnaz Sarlak
- INSERM U1211, 33000 Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Claude Lalou
- INSERM U1211, 33000 Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Nivea Dias Amoedo
- CELLOMET, Functional Genomics Center (CGFB), 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Rodrigue Rossignol
- INSERM U1211, 33000 Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33000 Bordeaux, France; CELLOMET, Functional Genomics Center (CGFB), 146 rue Léo Saignat, 33000 Bordeaux, France.
| |
Collapse
|
188
|
Arias E, Cuervo AM. Pros and Cons of Chaperone-Mediated Autophagy in Cancer Biology. Trends Endocrinol Metab 2020; 31:53-66. [PMID: 31699565 PMCID: PMC7020649 DOI: 10.1016/j.tem.2019.09.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 11/18/2022]
Abstract
Autophagy contributes to cellular quality control and energetics through lysosomal breakdown and recycling of essential cellular components. Chaperone-mediated autophagy (CMA) adds to these autophagic functions the ability to timely and selectively degrade single tagged proteins to terminate their cellular function and, in this way, participate in the regulation of multiple cellular processes. Many cancer cells upregulate CMA for protumorigenic and prosurvival purposes. However, growing evidence supports a physiological role for CMA in limiting malignant transformation. Understanding the mechanisms behind this functional switch of CMA from antioncogenic to pro-oncogenic is fundamental for targeting CMA in cancer treatment. We summarize current understanding of CMA functions in cancer biology and discuss the basis for its context-dependent dual role in oncogenesis.
Collapse
Affiliation(s)
- Esperanza Arias
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Ana Maria Cuervo
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
189
|
Kim E, Park S, Lee JH, Mun JY, Choi WH, Yun Y, Lee J, Kim JH, Kang MJ, Lee MJ. Dual Function of USP14 Deubiquitinase in Cellular Proteasomal Activity and Autophagic Flux. Cell Rep 2019; 24:732-743. [PMID: 30021169 DOI: 10.1016/j.celrep.2018.06.058] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/15/2018] [Accepted: 06/14/2018] [Indexed: 12/17/2022] Open
Abstract
The ubiquitin-proteasome system and the autophagy-lysosome system are two major intracellular proteolytic pathways in eukaryotes. Although several biochemical mechanisms underlying the crosstalk between them have been suggested, little is known about the effect of enhanced proteasome activity on autophagic flux. Here, we found that upregulation of proteasome activity, which was achieved through the inhibition of USP14, significantly impaired cellular autophagic flux, especially at the autophagosome-lysosome fusion step. UVRAG appeared to function as a crucial checkpoint for the proper progression of autophagic flux. Although proteasome activation through USP14 inhibition facilitated the clearance of microtubule-associated protein tau (MAPT) and reduced the amount of its oligomeric forms, the same conditions increased the formation of inclusion bodies from nonproteasomal substrates such as huntingtin with long polyglutamine repeats. Our results collectively indicate that USP14 may function as a common denominator in the compensatory negative feedback between the two major proteolytic processes in the cell.
Collapse
Affiliation(s)
- Eunkyoung Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Seoyoung Park
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jung Hoon Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ji Young Mun
- Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu 41068, Korea
| | - Won Hoon Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Yejin Yun
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Jeeyoung Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Ji Hyeon Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Min-Ji Kang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.
| | - Min Jae Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea; Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
190
|
Zhang X, Gaetani M, Chernobrovkin A, Zubarev RA. Anticancer Effect of Deuterium Depleted Water - Redox Disbalance Leads to Oxidative Stress. Mol Cell Proteomics 2019; 18:2373-2387. [PMID: 31519768 PMCID: PMC6885702 DOI: 10.1074/mcp.ra119.001455] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/22/2019] [Indexed: 12/31/2022] Open
Abstract
Despite the convincing empirical evidence that deuterium depleted water (DDW, 25-125 ppm deuterium) has anticancer effect, the molecular mechanism remains unclear. Here, redox proteomics investigation of the DDW action in A549 cells revealed an increased level of oxidative stress, whereas expression proteomics in combination with thermal profiling uncovered crucial role of mitochondrial proteins. In the proposed scenario, reversal of the normally positive deuterium gradient across the inner membrane leads to an increased export of protons from the matrix to intermembrane space and an increase in the mitochondrial membrane potential, enhancing the production of reactive oxygen species (ROS). The resulting oxidative stress leads to slower growth and can induce apoptosis. However, further deuterium depletion in ambient water triggers a feedback mechanism, which leads to restoration of the redox equilibrium and resumed growth. The DDW-induced oxidative stress, verified by traditional biochemical assays, may be helpful as an adjuvant to ROS-inducing anticancer therapy.
Collapse
Affiliation(s)
- Xuepei Zhang
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17 165 Stockholm, Sweden
| | - Massimiliano Gaetani
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17 165 Stockholm, Sweden
| | - Alexey Chernobrovkin
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17 165 Stockholm, Sweden
| | - Roman A Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17 165 Stockholm, Sweden.
| |
Collapse
|
191
|
Gao P, Zhang H, Zhang Q, Fang X, Wu H, Wang M, Lu Z, Wei X, Yang G, Yan Z, Liu D, Zhu Z. Caloric Restriction Exacerbates Angiotensin II-Induced Abdominal Aortic Aneurysm in the Absence of p53. Hypertension 2019; 73:547-560. [PMID: 30686087 DOI: 10.1161/hypertensionaha.118.12086] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
p53-dependent vascular smooth muscle cell senescence is a key pathological process of abdominal aortic aneurysm (AAA). Caloric restriction (CR) is a nonpharmacological intervention that prevents AAA formation. However, whether p53 is indispensable to the protective role of CR remains unknown. In this study, we investigated the necessity of p53 in the beneficial role of CR in AAA formation and the underlying mechanisms. We subjected p53+/+ and p53-/- mice to 12 weeks of CR and then examined the incidence of Ang II (angiotensin II)-induced AAA formation. We found that both CR and p53 knockout reduced Ang II-induced AAA formation; however, CR markedly increased the incidence of AAA formation and exacerbated aortic elastin degradation in p53-/- mice, accompanied by increased vascular senescence, reactive oxygen species generation, and reduced energy production. Analysis of mitochondrial respiratory activity revealed that dysfunctional complex IV accounts for the abnormal mitochondrial respiration in p53-/- vascular smooth muscle cells treated by CR serum. Mechanistically, ablation of p53 almost totally blocked the protective role of CR by inhibiting SCO2 (cytochrome C oxidase assembly protein 2)-dependent mitochondrial complex IV activity. Overexpression of SCO2 restored the beneficial effect of CR on antagonizing Ang II-induced expression of AAA-related molecules and reactive oxygen species generation in p53-/- vascular smooth muscle cells. Together, our findings demonstrate that the existence of p53 in vascular smooth muscle cells is critical to the protective role of CR in Ang II-induced AAA formation by maintaining an appropriate mitochondrial function.
Collapse
Affiliation(s)
- Peng Gao
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, China (P.G., H.Z., H.W., Z.L., X.W., Z.Y., D.L., Z.Z.)
| | - Hexuan Zhang
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, China (P.G., H.Z., H.W., Z.L., X.W., Z.Y., D.L., Z.Z.)
| | - Qin Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University and Chongqing Clinical Research Center for Geriatrics, China (Q.Z., X.F., M.W., G.Y.)
| | - Xia Fang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University and Chongqing Clinical Research Center for Geriatrics, China (Q.Z., X.F., M.W., G.Y.)
| | - Hao Wu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, China (P.G., H.Z., H.W., Z.L., X.W., Z.Y., D.L., Z.Z.)
| | - Miao Wang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University and Chongqing Clinical Research Center for Geriatrics, China (Q.Z., X.F., M.W., G.Y.)
| | - Zongshi Lu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, China (P.G., H.Z., H.W., Z.L., X.W., Z.Y., D.L., Z.Z.)
| | - Xiao Wei
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, China (P.G., H.Z., H.W., Z.L., X.W., Z.Y., D.L., Z.Z.)
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University and Chongqing Clinical Research Center for Geriatrics, China (Q.Z., X.F., M.W., G.Y.)
| | - Zhencheng Yan
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, China (P.G., H.Z., H.W., Z.L., X.W., Z.Y., D.L., Z.Z.)
| | - Daoyan Liu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, China (P.G., H.Z., H.W., Z.L., X.W., Z.Y., D.L., Z.Z.)
| | - Zhiming Zhu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, China (P.G., H.Z., H.W., Z.L., X.W., Z.Y., D.L., Z.Z.)
| |
Collapse
|
192
|
The Rich World of p53 DNA Binding Targets: The Role of DNA Structure. Int J Mol Sci 2019; 20:ijms20225605. [PMID: 31717504 PMCID: PMC6888028 DOI: 10.3390/ijms20225605] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 10/29/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor suppressor functions of p53 and its roles in regulating the cell cycle, apoptosis, senescence, and metabolism are accomplished mainly by its interactions with DNA. p53 works as a transcription factor for a significant number of genes. Most p53 target genes contain so-called p53 response elements in their promoters, consisting of 20 bp long canonical consensus sequences. Compared to other transcription factors, which usually bind to one concrete and clearly defined DNA target, the p53 consensus sequence is not strict, but contains two repeats of a 5′RRRCWWGYYY3′ sequence; therefore it varies remarkably among target genes. Moreover, p53 binds also to DNA fragments that at least partially and often completely lack this consensus sequence. p53 also binds with high affinity to a variety of non-B DNA structures including Holliday junctions, cruciform structures, quadruplex DNA, triplex DNA, DNA loops, bulged DNA, and hemicatenane DNA. In this review, we summarize information of the interactions of p53 with various DNA targets and discuss the functional consequences of the rich world of p53 DNA binding targets for its complex regulatory functions.
Collapse
|
193
|
ASPP2 inhibits tumor growth by repressing the mevalonate pathway in hepatocellular carcinoma. Cell Death Dis 2019; 10:830. [PMID: 31685796 PMCID: PMC6828733 DOI: 10.1038/s41419-019-2054-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 09/18/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022]
Abstract
Cancer is, fundamentally, a disorder of cell growth and proliferation, which requires adequate supplies of energy and nutrients. In this study, we report that the haplo-insufficient tumor suppressor ASPP2, a p53 activator, negatively regulates the mevalonate pathway to mediate its inhibitory effect on tumor growth in hepatocellular carcinoma (HCC). Gene expression profile analysis revealed that the expression of key enzymes in the mevalonate pathway were increased when ASPP2 was downregulated. HCC cells gained higher cholesterol levels and enhanced tumor-initiating capability in response to the depletion of ASPP2. Simvastatin, a mevalonate pathway inhibitor, efficiently abrogated ASPP2 depletion-induced anchorage-independent cell proliferation, resistance to chemotherapy drugs in vitro, and tumor growth in xenografted nude mice. Mechanistically, ASPP2 interacts with SREBP-2 in the nucleus and restricts the transcriptional activity of SREBP-2 on its target genes, which include key enzymes involved in the mevalonate pathway. Moreover, clinical data revealed better prognosis in patients with high levels of ASPP2 and low levels of the mevalonate pathway enzyme HMGCR. Our findings provide functional and mechanistic insights into the critical role of ASPP2 in the regulation of the mevalonate pathway and the importance of this pathway in tumor initiation and tumor growth, which may provide a new therapeutic opportunity for HCC.
Collapse
|
194
|
Kim E, Kim JY, Lee JY. Mathematical Modeling of p53 Pathways. Int J Mol Sci 2019; 20:ijms20205179. [PMID: 31635420 PMCID: PMC6834204 DOI: 10.3390/ijms20205179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/15/2019] [Accepted: 10/15/2019] [Indexed: 12/30/2022] Open
Abstract
Cells have evolved balanced systems that ensure an appropriate response to stress. The systems elicit repair responses in temporary or moderate stress but eliminate irreparable cells via apoptosis in detrimental conditions of prolonged or severe stress. The tumor suppressor p53 is a central player in these stress response systems. When activated under DNA damage stress, p53 regulates hundreds of genes that are involved in DNA repair, cell cycle, and apoptosis. Recently, increasing studies have demonstrated additional regulatory roles of p53 in metabolism and mitochondrial physiology. Due to the inherent complexity of feedback loops between p53 and its target genes, the application of mathematical modeling has emerged as a novel approach to better understand the multifaceted functions and dynamics of p53. In this review, we discuss several mathematical modeling approaches in exploring the p53 pathways.
Collapse
Affiliation(s)
- Eunjung Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Korea.
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Korea.
- Korea Basic Science Institute, Daejeon 34133, Korea.
| | - Joo-Yong Lee
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Korea.
- Korea Basic Science Institute, Daejeon 34133, Korea.
| |
Collapse
|
195
|
Rodríguez-Enríquez S, Marín-Hernández Á, Gallardo-Pérez JC, Pacheco-Velázquez SC, Belmont-Díaz JA, Robledo-Cadena DX, Vargas-Navarro JL, Corona de la Peña NA, Saavedra E, Moreno-Sánchez R. Transcriptional Regulation of Energy Metabolism in Cancer Cells. Cells 2019; 8:cells8101225. [PMID: 31600993 PMCID: PMC6830338 DOI: 10.3390/cells8101225] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/19/2019] [Accepted: 10/01/2019] [Indexed: 01/17/2023] Open
Abstract
Cancer development, growth, and metastasis are highly regulated by several transcription regulators (TRs), namely transcription factors, oncogenes, tumor-suppressor genes, and protein kinases. Although TR roles in these events have been well characterized, their functions in regulating other important cancer cell processes, such as metabolism, have not been systematically examined. In this review, we describe, analyze, and strive to reconstruct the regulatory networks of several TRs acting in the energy metabolism pathways, glycolysis (and its main branching reactions), and oxidative phosphorylation of nonmetastatic and metastatic cancer cells. Moreover, we propose which possible gene targets might allow these TRs to facilitate the modulation of each energy metabolism pathway, depending on the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Norma Angélica Corona de la Peña
- Unidad de Investigación Médica en Trombosis, Hemostasia y Aterogénesis, Hospital General Regional Carlos McGregor-Sánchez, México CP 03100, Mexico.
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología, México 14080, Mexico.
| | - Rafael Moreno-Sánchez
- Departamento de Bioquímica, Instituto Nacional de Cardiología, México 14080, Mexico.
| |
Collapse
|
196
|
Zhu S, Corsetti S, Wang Q, Li C, Huang Z, Nabi G. Optical sensory arrays for the detection of urinary bladder cancer-related volatile organic compounds. JOURNAL OF BIOPHOTONICS 2019; 12:e201800165. [PMID: 30168296 PMCID: PMC7065633 DOI: 10.1002/jbio.201800165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 08/29/2018] [Indexed: 05/06/2023]
Abstract
Non-invasive detection of urinary bladder cancer remains a significant challenge. Urinary volatile organic compounds (VOCs) are a promising alternative to cell-based biomarkers. Herein, we demonstrate a novel diagnosis system based on an optic fluorescence sensor array for detecting urinary bladder cancer VOCs biomarkers. This study describes a fluorescence-based VOCs sensor array detecting system in detail. The choice of VOCs for the initial part was based on an extensive systematic search of the literature and then followed up using urinary samples from patients with urinary bladder transitional cell carcinoma. Canonical discriminant analysis and partial least squares discriminant analysis (PLS-DA) were employed and correctly detected 31/48 urinary bladder cancer VOC biomarkers and achieved an overall 77.75% sensitivity and 93.25% specificity by PLS-DA modelling. All five urine samples from bladder cancer patients, and five healthy controls were successfully identified with the same sensor arrays. Overall, the experiments in this study describe a real-time platform for non-invasive bladder cancer diagnosis using fluorescence-based gas-sensor arrays. Pure VOCs and urine samples from the patients proved such a system to be promising; however, further research is required using a larger population sample.
Collapse
Affiliation(s)
- Simian Zhu
- Cancer Research DivisionSchool of Medicine, University of DundeeDundeeUK
- Mechanical and Electronic Engineering, School of Science and EngineeringUniversity of DundeeDundeeUK
| | | | - Qifan Wang
- Mechanical and Electronic Engineering, School of Science and EngineeringUniversity of DundeeDundeeUK
| | - Chunhui Li
- Mechanical and Electronic Engineering, School of Science and EngineeringUniversity of DundeeDundeeUK
| | - Zhihong Huang
- Mechanical and Electronic Engineering, School of Science and EngineeringUniversity of DundeeDundeeUK
| | - Ghulam Nabi
- Cancer Research DivisionSchool of Medicine, University of DundeeDundeeUK
| |
Collapse
|
197
|
Njomen E, Tepe JJ. Regulation of Autophagic Flux by the 20S Proteasome. Cell Chem Biol 2019; 26:1283-1294.e5. [PMID: 31327703 PMCID: PMC6754308 DOI: 10.1016/j.chembiol.2019.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/24/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022]
Abstract
The proteolytic arm of the protein homeostasis network is maintained by both the ubiquitin-proteasome system (UPS) and autophagy. A well-balanced crosstalk between the two catabolic pathways ensures energy-efficient maintenance of cellular function. Our current understanding of the crosstalk between the UPS and autophagy is centered around substrate ubiquitination. Herein we report an additional method of crosstalk involving ubiquitin-independent 20S proteasome regulation of autophagosome-lysosome fusion. We found that enhancement of 20S proteasome activity increased the degradation of the disordered soluble N-ethylmaleimide-sensitive factor activating protein receptor proteins, synaptosomal-associated protein 29 (SNAP29), and syntaxin 17 (STX17), but not vesicle-associated membrane protein 8. This resulted in a reduction of autophagosome-lysosome fusion, which was ameliorated upon overexpression of both SNAP29 and STX17. In all, we herein present a mechanism of crosstalk between the proteasome and autophagy pathway that is regulated by ubiquitin-independent 20S proteasome-mediated degradation of SNAP29 and STX17.
Collapse
Affiliation(s)
- Evert Njomen
- Department of Chemistry and Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Jetze J Tepe
- Department of Chemistry and Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
198
|
Han CY, Patten DA, Lee SG, Parks RJ, Chan DW, Harper M, Tsang BK. p53 Promotes chemoresponsiveness by regulating hexokinase II gene transcription and metabolic reprogramming in epithelial ovarian cancer. Mol Carcinog 2019; 58:2161-2174. [DOI: 10.1002/mc.23106] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 08/16/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Chae Young Han
- Department of Obstetrics & Gynecology and Cellular & Molecular MedicineUniversity of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute Ottawa Ontario Canada
| | - David A. Patten
- Department of Biochemistry, Microbiology and ImmunologyUniversity of Ottawa, Institute of Systems Biology Ottawa Ontario Canada
| | - Seung Gee Lee
- Department of Obstetrics & Gynecology and Cellular & Molecular MedicineUniversity of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute Ottawa Ontario Canada
| | - Robin J. Parks
- Department of Biochemistry, Microbiology & ImmunologyUniversity of Ottawa and Regenerative Medicine Program, Ottawa Hospital Research Institute Ottawa Ontario Canada
| | - David W. Chan
- Department of Obstetrics and GynecologyThe University of Hong Kong Hong Kong SAR P.R. China
| | - Mary‐Ellen Harper
- Department of Biochemistry, Microbiology and ImmunologyUniversity of Ottawa, Institute of Systems Biology Ottawa Ontario Canada
| | - Benjamin K. Tsang
- Department of Obstetrics & Gynecology and Cellular & Molecular MedicineUniversity of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute Ottawa Ontario Canada
| |
Collapse
|
199
|
Yu L, Yu TT, Young KH. Cross-talk between Myc and p53 in B-cell lymphomas. Chronic Dis Transl Med 2019; 5:139-154. [PMID: 31891126 PMCID: PMC6926120 DOI: 10.1016/j.cdtm.2019.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Indexed: 02/07/2023] Open
Abstract
Myc and p53 proteins are closely associated with many physiological cellular functions, including immune response and lymphocyte survival, and are expressed in the lymphoid organs, which are sites for the development and activation of B-cell malignancies. Genetic alterations and other mechanisms resulting in constitutive activation, rearrangement, or mutation of MYC and TP53 contribute to the development of lymphomas, progression and therapy resistance by gene dysregulation, activation of downstream anti-apoptotic pathways, and unfavorable microenvironment interactions. The cross-talk between the Myc and p53 proteins contributes to the inferior prognosis in many types of B-cell lymphomas. In this review, we present the physiological roles of Myc and p53 proteins, and recent advances in understanding the pathological roles of Myc, p53, and their cross-talk in lymphoid neoplasms. In addition, we highlight clinical trials of novel agents that directly or indirectly inhibit Myc and/or p53 protein functions and their signaling pathways. Although, to date, these trials have failed to overcome drug resistance, the new results have highlighted the clinical efficiency of targeting diverse mechanisms of action with the goal of optimizing novel therapeutic opportunities to eradicate lymphoma cells.
Collapse
Affiliation(s)
- Li Yu
- Department of Hematology, The Second Affiliated Hospital to Nanchang University, Nanchang, Jiangxi 330006, China
- Hematopathology Division and Pathology Department, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tian-Tian Yu
- Department of Hematology, The Second Affiliated Hospital to Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ken H. Young
- Hematopathology Division and Pathology Department, Duke University School of Medicine, Durham, NC 27710, USA
- Duke University Medical Center and Cancer Institute, Durham, NC 27710, USA
| |
Collapse
|
200
|
Yang H, Shu Z, Jiang Y, Mao W, Pang L, Redwood A, Jeter-Jones SL, Jennings NB, Ornelas A, Zhou J, Rodriguez-Aguayo C, Bartholomeusz G, Iles LR, Zacharias NM, Millward SW, Lopez-Berestein G, Le XF, Ahmed AA, Piwnica-Worms H, Sood AK, Bast RC, Lu Z. 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase-2 Regulates TP53-Dependent Paclitaxel Sensitivity in Ovarian and Breast Cancers. Clin Cancer Res 2019; 25:5702-5716. [PMID: 31391192 DOI: 10.1158/1078-0432.ccr-18-3448] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/21/2019] [Accepted: 06/13/2019] [Indexed: 01/23/2023]
Abstract
PURPOSE Paclitaxel is an integral component of primary therapy for breast and epithelial ovarian cancers, but less than half of these cancers respond to the drug. Enhancing the response to primary therapy with paclitaxel could improve outcomes for women with both diseases.Experimental Design: Twelve kinases that regulate metabolism were depleted in multiple ovarian and breast cancer cell lines to determine whether they regulate sensitivity to paclitaxel in Sulforhodamine B assays. The effects of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 2 (PFKFB2) depletion on cell metabolomics, extracellular acidification rate, nicotinamide adenine dinucleotide phosphate, reactive oxygen species (ROS), and apoptosis were studied in multiple ovarian and breast cancer cell lines. Four breast and ovarian human xenografts and a breast cancer patient-derived xenograft (PDX) were used to examine the knockdown effect of PFKFB2 on tumor cell growth in vivo. RESULTS Knockdown of PFKFB2 inhibited clonogenic growth and enhanced paclitaxel sensitivity in ovarian and breast cancer cell lines with wild-type TP53 (wtTP53). Silencing PFKFB2 significantly inhibited tumor growth and enhanced paclitaxel sensitivity in four xenografts derived from two ovarian and two breast cancer cell lines, and prolonged survival in a triple-negative breast cancer PDX. Transfection of siPFKFB2 increased the glycolysis rate, but decreased the flow of intermediates through the pentose-phosphate pathway in cancer cells with wtTP53, decreasing NADPH. ROS accumulated after PFKFB2 knockdown, which stimulated Jun N-terminal kinase and p53 phosphorylation, and induced apoptosis that depended upon upregulation of p21 and Puma. CONCLUSIONS PFKFB2 is a novel target whose inhibition can enhance the effect of paclitaxel-based primary chemotherapy upon ovarian and breast cancers retaining wtTP53.
Collapse
Affiliation(s)
- Hailing Yang
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Zhang Shu
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas.,Department of Geriatric Digestive Surgery, the Second Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yongying Jiang
- Institute for Applied Cancer Science, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Weiqun Mao
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Lan Pang
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Abena Redwood
- Department of Experimental Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Sabrina L Jeter-Jones
- Department of Experimental Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Nicholas B Jennings
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Argentina Ornelas
- Cancer Systems Imaging, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Jinhua Zhou
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas.,Center for RNA Interference and Non-Coding RNA Cancer, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Geoffrey Bartholomeusz
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - LaKesla R Iles
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Niki M Zacharias
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Steven W Millward
- Cancer Systems Imaging, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas.,Center for RNA Interference and Non-Coding RNA Cancer, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Xiao-Feng Le
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Ahmed A Ahmed
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, United Kingdom.,Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, Houston, Texas.,Center for RNA Interference and Non-Coding RNA Cancer, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Robert C Bast
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas.
| | - Zhen Lu
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|