1
|
Glorieux C, Buc Calderon P. Targeting catalase in cancer. Redox Biol 2024; 77:103404. [PMID: 39447253 DOI: 10.1016/j.redox.2024.103404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Healthy cells have developed a sophisticated network of antioxidant molecules to prevent the toxic accumulation of reactive oxygen species (ROS) generated by diverse environmental stresses. On the opposite, cancer cells often exhibit high levels of ROS and an altered levels of antioxidant molecules compared to normal cells. Among them, the antioxidant enzyme catalase plays an essential role in cell defense against oxidative stress through the dismutation of hydrogen peroxide into water and molecular oxygen, and its expression is often decreased in cancer cells. The elevation of ROS in cancer cells provides them proliferative advantages, and leads to metabolic reprogramming, immune escape and metastasis. In this context, catalase is of critical importance to control these cellular processes in cancer through various mechanisms. In this review, we will discuss the major progresses and challenges in understanding the role of catalase in cancer for this last decade. This review also aims to provide important updates regarding the regulation of catalase expression, subcellular localization and discuss about the potential role of microbial catalases in tumor environment. Finally, we will describe the different catalase-based therapies and address the advantages, disadvantages, and limitations associated with modulating catalase therapeutically in cancer treatment.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060, Guangzhou, China.
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de La Salud, Universidad Arturo Prat, 1100000, Iquique, Chile; Instituto de Química Medicinal, Universidad Arturo Prat, 1100000, Iquique, Chile; Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université Catholique de Louvain, 1200, Brussels, Belgium.
| |
Collapse
|
2
|
Nagano S, Unuma K, Aki T, Uemura K. N-acetylcysteine alleviates arsenic trioxide-induced reductions in hepatic catalase gene expression both in vitro and in vivo. Leg Med (Tokyo) 2024; 69:102458. [PMID: 38781725 DOI: 10.1016/j.legalmed.2024.102458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Arsenic trioxide (ATO), one of the oldest and most frequently used poisons, is well-known in forensic science for inducing hepatotoxicity. The regulation of peroxisomal antioxidative enzyme catalase (CAT) involves intricate mechanisms at both transcriptional and post-transcriptional levels. However, the molecular mechanisms underlying the regulation of CAT gene expression in hepatic cells remain elusive. Furthermore, the regulation of CAT gene expression evident in animals administered with ATO in vivo is not well-explored, although several studies have revealed ATO-induced reductions in CAT enzymatic activity in rat livers. In this study, we revealed ATO-dependent reductions in CAT gene expression in both rat liver and Huh-7 human hepatoma cells. Our results indicate that the decline in CAT enzymatic activity can be attributed, at least in part, to the downregulation of its gene expression. The ATO-induced reduction in CAT expression was concurrent with the reduction in peroxisome proliferator-activated receptor-gamma (PPARγ) coactivator (PGC)-1α and inactivation of PPARγ, both considered as positive regulators of CAT gene expression. Moreover, antioxidant N-acetylcysteine (NAC) demonstrated the capability to alleviate the downregulation of CAT gene expression both in vivo and in vitro. Additionally, NAC played a role in alleviating ATO-induced hepatotoxicity, potentially by mitigating the transcriptional downregulation of the CAT gene. Altogether, these results indicate that ATO exerts toxicity by inhibiting the antioxidant defense mechanism, which may be useful for forensic diagnosis of arsenic poisoning and clinical treatment of mitigating ATO-induced hepatotoxicity.
Collapse
Affiliation(s)
- Shutaro Nagano
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
3
|
Rainey NE, Armand AS, Petit PX. Sodium arsenite and arsenic trioxide differently affect the oxidative stress of lymphoblastoid cells: An intricate crosstalk between mitochondria, autophagy and cell death. PLoS One 2024; 19:e0302701. [PMID: 38728286 PMCID: PMC11086853 DOI: 10.1371/journal.pone.0302701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Although the toxicity of arsenic depends on its chemical forms, few studies have taken into account the ambiguous phenomenon that sodium arsenite (NaAsO2) acts as a potent carcinogen while arsenic trioxide (ATO, As2O3) serves as an effective therapeutic agent in lymphoma, suggesting that NaAsO2 and As2O3 may act via paradoxical ways to either promote or inhibit cancer pathogenesis. Here, we compared the cellular response of the two arsenical compounds, NaAsO2 and As2O3, on the Burkitt lymphoma cell model, the Epstein Barr Virus (EBV)-positive P3HR1 cells. Using flow cytometry and biochemistry analyses, we showed that a NaAsO2 treatment induces P3HR1 cell death, combined with drastic drops in ΔΨm, NAD(P)H and ATP levels. In contrast, As2O3-treated cells resist to cell death, with a moderate reduction of ΔΨm, NAD(P)H and ATP. While both compounds block cells in G2/M and affect their protein carbonylation and lipid peroxidation, As2O3 induces a milder increase in superoxide anions and H2O2 than NaAsO2, associated to a milder inhibition of antioxidant defenses. By electron microscopy, RT-qPCR and image cytometry analyses, we showed that As2O3-treated cells display an overall autophagic response, combined with mitophagy and an unfolded protein response, characteristics that were not observed following a NaAsO2 treatment. As previous works showed that As2O3 reactivates EBV in P3HR1 cells, we treated the EBV- Ramos-1 cells and showed that autophagy was not induced in these EBV- cells upon As2O3 treatment suggesting that the boost of autophagy observed in As2O3-treated P3HR1 cells could be due to the presence of EBV in these cells. Overall, our results suggest that As2O3 is an autophagic inducer which action is enhanced when EBV is present in the cells, in contrast to NaAsO2, which induces cell death. That's why As2O3 is combined with other chemicals, as all-trans retinoic acid, to better target cancer cells in therapeutic treatments.
Collapse
Affiliation(s)
- Nathan Earl Rainey
- CNRS UMR 8003 Paris University, SSPIN, Neuroscience Institute, Team “Mitochondria, Apoptosis and Autophagy Signaling”, Campus Saint-Germain, Paris, France
| | - Anne-Sophie Armand
- INSERM U1151, Institut Necker Enfants Malades (INEM), Campus Necker, Université Paris Cité, Paris, France
| | - Patrice X. Petit
- CNRS UMR 8003 Paris University, SSPIN, Neuroscience Institute, Team “Mitochondria, Apoptosis and Autophagy Signaling”, Campus Saint-Germain, Paris, France
| |
Collapse
|
4
|
Jomova K, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, Valko M. Several lines of antioxidant defense against oxidative stress: antioxidant enzymes, nanomaterials with multiple enzyme-mimicking activities, and low-molecular-weight antioxidants. Arch Toxicol 2024; 98:1323-1367. [PMID: 38483584 PMCID: PMC11303474 DOI: 10.1007/s00204-024-03696-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 01/31/2024] [Indexed: 03/27/2024]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are well recognized for playing a dual role, since they can be either deleterious or beneficial to biological systems. An imbalance between ROS production and elimination is termed oxidative stress, a critical factor and common denominator of many chronic diseases such as cancer, cardiovascular diseases, metabolic diseases, neurological disorders (Alzheimer's and Parkinson's diseases), and other disorders. To counteract the harmful effects of ROS, organisms have evolved a complex, three-line antioxidant defense system. The first-line defense mechanism is the most efficient and involves antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). This line of defense plays an irreplaceable role in the dismutation of superoxide radicals (O2•-) and hydrogen peroxide (H2O2). The removal of superoxide radicals by SOD prevents the formation of the much more damaging peroxynitrite ONOO- (O2•- + NO• → ONOO-) and maintains the physiologically relevant level of nitric oxide (NO•), an important molecule in neurotransmission, inflammation, and vasodilation. The second-line antioxidant defense pathway involves exogenous diet-derived small-molecule antioxidants. The third-line antioxidant defense is ensured by the repair or removal of oxidized proteins and other biomolecules by a variety of enzyme systems. This review briefly discusses the endogenous (mitochondria, NADPH, xanthine oxidase (XO), Fenton reaction) and exogenous (e.g., smoking, radiation, drugs, pollution) sources of ROS (superoxide radical, hydrogen peroxide, hydroxyl radical, peroxyl radical, hypochlorous acid, peroxynitrite). Attention has been given to the first-line antioxidant defense system provided by SOD, CAT, and GPx. The chemical and molecular mechanisms of antioxidant enzymes, enzyme-related diseases (cancer, cardiovascular, lung, metabolic, and neurological diseases), and the role of enzymes (e.g., GPx4) in cellular processes such as ferroptosis are discussed. Potential therapeutic applications of enzyme mimics and recent progress in metal-based (copper, iron, cobalt, molybdenum, cerium) and nonmetal (carbon)-based nanomaterials with enzyme-like activities (nanozymes) are also discussed. Moreover, attention has been given to the mechanisms of action of low-molecular-weight antioxidants (vitamin C (ascorbate), vitamin E (alpha-tocopherol), carotenoids (e.g., β-carotene, lycopene, lutein), flavonoids (e.g., quercetin, anthocyanins, epicatechin), and glutathione (GSH)), the activation of transcription factors such as Nrf2, and the protection against chronic diseases. Given that there is a discrepancy between preclinical and clinical studies, approaches that may result in greater pharmacological and clinical success of low-molecular-weight antioxidant therapies are also subject to discussion.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine The Philosopher University in Nitra, Nitra, 949 74, Slovakia
| | - Suliman Y Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Saleh H Alwasel
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
5
|
Chien TM, Yang CW, Yen CH, Yeh BW, Wu WJ, Sheu JH, Chang HW. Excavatolide C/cisplatin combination induces antiproliferation and drives apoptosis and DNA damage in bladder cancer cells. Arch Toxicol 2024; 98:1543-1560. [PMID: 38424264 DOI: 10.1007/s00204-024-03699-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
Excavatolide C (EXCC), a marine coral-derived compound, exhibits an antiproliferation effect on bladder cancer cells. The present study evaluated the improvement in the antiproliferation ability of EXCC by co-treatment with cisplatin in bladder cancer cells. EXCC/cisplatin (12.5 and 1 μg/mL) showed higher antiproliferation effects on bladder cancer cells than single treatments (EXCC or cisplatin alone) in the 48 h ATP assay. EXCC/cisplatin also enhanced the increase in subG1, annexin V-mediated apoptosis, and activation of poly (ADP-ribose) polymerase (PARP) and several caspases (caspases 3, 8, and 9) compared to the single treatments. Cellular and mitochondrial oxidative stress was enhanced with EXCC/cisplatin compared to the single treatments according to analyses of reactive oxygen species (ROS), mitochondrial superoxide, and mitochondrial membrane potential; in addition, cellular antioxidants, such as glutathione (GSH), and the mRNA expressions of antioxidant signaling genes (catalase and NFE2-like bZIP transcription factor 2) were downregulated. EXCC/cisplatin treatment produced more DNA damage than the single treatments, as indicated by γH2AX and 8-hydroxy-2'-deoxyguanosine levels. Moreover, several DNA repair genes for homologous recombination (HR) and non-homologous end joining (NHEJ) were downregulated in EXCC/cisplatin compared to others. The addition of the GSH precursor N-acetylcysteine, which has ROS scavenging activity, attenuated all EXCC/cisplatin-induced changes. Notably, EXCC/cisplatin showed lower antiproliferation, apoptosis, ROS induction, GSH depletion, and γH2AX DNA damage in normal cells than in bladder cancer cells. Therefore, the co-treatment of EXCC/cisplatin reduces the proliferation of bladder cancer cells via oxidative stress-mediated mechanisms with normal cell safety.
Collapse
Affiliation(s)
- Tsu-Ming Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
- Department of Urology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Che-Wei Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Bi-Wen Yeh
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
| | - Wen-Jeng Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
- Department of Urology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Jyh-Horng Sheu
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan.
| | - Hsueh-Wei Chang
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
6
|
Rasheed Z. Therapeutic potentials of catalase: Mechanisms, applications, and future perspectives. Int J Health Sci (Qassim) 2024; 18:1-6. [PMID: 38455600 PMCID: PMC10915913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024] Open
Affiliation(s)
- Zafar Rasheed
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| |
Collapse
|
7
|
Glorieux C, Enríquez C, González C, Aguirre-Martínez G, Buc Calderon P. The Multifaceted Roles of NRF2 in Cancer: Friend or Foe? Antioxidants (Basel) 2024; 13:70. [PMID: 38247494 PMCID: PMC10812565 DOI: 10.3390/antiox13010070] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Physiological concentrations of reactive oxygen species (ROS) play vital roles in various normal cellular processes, whereas excessive ROS generation is central to disease pathogenesis. The nuclear factor erythroid 2-related factor 2 (NRF2) is a critical transcription factor that regulates the cellular antioxidant systems in response to oxidative stress by governing the expression of genes encoding antioxidant enzymes that shield cells from diverse oxidative alterations. NRF2 and its negative regulator Kelch-like ECH-associated protein 1 (KEAP1) have been the focus of numerous investigations in elucidating whether NRF2 suppresses tumor promotion or conversely exerts pro-oncogenic effects. NRF2 has been found to participate in various pathological processes, including dysregulated cell proliferation, metabolic remodeling, and resistance to apoptosis. Herein, this review article will examine the intriguing role of phase separation in activating the NRF2 transcriptional activity and explore the NRF2 dual impacts on tumor immunology, cancer stem cells, metastasis, and long non-coding RNAs (LncRNAs). Taken together, this review aims to discuss the NRF2 multifaceted roles in both cancer prevention and promotion while also addressing the advantages, disadvantages, and limitations associated with modulating NRF2 therapeutically in cancer treatment.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Cinthya Enríquez
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
- Programa de Magister en Ciencias Químicas y Farmacéuticas, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile
| | - Constanza González
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
| | - Gabriela Aguirre-Martínez
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
- Instituto de Química Medicinal, Universidad Arturo Prat, Iquique 1100000, Chile
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
- Instituto de Química Medicinal, Universidad Arturo Prat, Iquique 1100000, Chile
- Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
8
|
Hassan HA, Ahmed HS, Hassan DF. Free radicals and oxidative stress: Mechanisms and therapeutic targets. Hum Antibodies 2024; 32:151-167. [PMID: 39031349 DOI: 10.3233/hab-240011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2024]
Abstract
BACKGROUND Free radicals are small extremely reactive species that have unpaired electrons. Free radicals include subgroups of reactive species, which are all a product of regular cellular metabolism. Oxidative stress happens when the free radicals production exceeds the capacity of the antioxidant system in the body's cells. OBJECTIVE The current review clarifies the prospective role of antioxidants in the inhibition and healing of diseases. METHODS Information on oxidative stress, free radicals, reactive oxidant species, and natural and synthetic antioxidants was obtained by searching electronic databases like PubMed, Web of Science, and Science Direct, with articles published between 1987 and 2023 being included in this review. RESULTS Free radicals exhibit a dual role in living systems. They are toxic byproducts of aerobic metabolism that lead to oxidative injury and tissue disorders and act as signals to activate appropriate stress responses. Endogenous and exogenous sources of reactive oxygen species are discussed in this review. Oxidative stress is a component of numerous diseases, including diabetes mellitus, atherosclerosis, cardiovascular disease, Alzheimer's disease, Parkinson's disease, and cancer. Although various small molecules assessed as antioxidants have shown therapeutic prospects in preclinical studies, clinical trial outcomes have been inadequate. Understanding the mechanisms through which antioxidants act, where, and when they are active may reveal a rational approach that leads to more tremendous pharmacological success. This review studies the associations between oxidative stress, redox signaling, and disease, the mechanisms through which oxidative stress can donate to pathology, the antioxidant defenses, the limits of their effectiveness, and antioxidant defenses that can be increased through physiological signaling, dietary constituents, and probable pharmaceutical interference. Prospective clinical applications of enzyme mimics and current progress in metal- and non-metal-based materials with enzyme-like activities and protection against chronic diseases have been discussed. CONCLUSION This review discussed oxidative stress as one of the main causes of illnesses, as well as antioxidant systems and their defense mechanisms that can be useful in inhibiting these diseases. Thus, the positive and deleterious effects of antioxidant molecules used to lessen oxidative stress in numerous human diseases are discussed. The optimal level of vitamins and minerals is the amount that achieves the best feed benefit, best growth rate, and health, including immune efficiency, and provides sufficient amounts to the body.
Collapse
|
9
|
Galasso M, Dalla Pozza E, Chignola R, Gambino S, Cavallini C, Quaglia FM, Lovato O, Dando I, Malpeli G, Krampera M, Donadelli M, Romanelli MG, Scupoli MT. The rs1001179 SNP and CpG methylation regulate catalase expression in chronic lymphocytic leukemia. Cell Mol Life Sci 2022; 79:521. [PMID: 36112236 PMCID: PMC9481481 DOI: 10.1007/s00018-022-04540-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/23/2022] [Accepted: 08/28/2022] [Indexed: 11/26/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is an incurable disease characterized by an extremely variable clinical course. We have recently shown that high catalase (CAT) expression identifies patients with an aggressive clinical course. Elucidating mechanisms regulating CAT expression in CLL is preeminent to understand disease mechanisms and develop strategies for improving its clinical management. In this study, we investigated the role of the CAT promoter rs1001179 single nucleotide polymorphism (SNP) and of the CpG Island II methylation encompassing this SNP in the regulation of CAT expression in CLL. Leukemic cells harboring the rs1001179 SNP T allele exhibited a significantly higher CAT expression compared with cells bearing the CC genotype. CAT promoter harboring the T -but not C- allele was accessible to ETS-1 and GR-β transcription factors. Moreover, CLL cells exhibited lower methylation levels than normal B cells, in line with the higher CAT mRNA and protein expressed by CLL in comparison with normal B cells. Methylation levels at specific CpG sites negatively correlated with CAT levels in CLL cells. Inhibition of methyltransferase activity induced a significant increase in CAT levels, thus functionally validating the role of CpG methylation in regulating CAT expression in CLL. Finally, the CT/TT genotypes were associated with lower methylation and higher CAT levels, suggesting that the rs1001179 T allele and CpG methylation may interact in regulating CAT expression in CLL. This study identifies genetic and epigenetic mechanisms underlying differential expression of CAT, which could be of crucial relevance for the development of therapies targeting redox regulatory pathways in CLL.
Collapse
Affiliation(s)
- Marilisa Galasso
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
- Section of Hematology, Department of Medicine, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Elisa Dalla Pozza
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
| | - Roberto Chignola
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134, Verona, Italy
| | - Simona Gambino
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
| | - Chiara Cavallini
- Research Center LURM, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Francesca Maria Quaglia
- Section of Hematology, Department of Medicine, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Ornella Lovato
- Research Center LURM, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Ilaria Dando
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
| | - Giorgio Malpeli
- Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Mauro Krampera
- Section of Hematology, Department of Medicine, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Massimo Donadelli
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
| | - Maria G Romanelli
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy.
| | - Maria T Scupoli
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy.
- Research Center LURM, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy.
| |
Collapse
|
10
|
Murillo MI, Gaiddon C, Le Lagadec R. Targeting of the intracellular redox balance by metal complexes towards anticancer therapy. Front Chem 2022; 10:967337. [PMID: 36034648 PMCID: PMC9405673 DOI: 10.3389/fchem.2022.967337] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The development of cancers is often linked to the alteration of essential redox processes, and therefore, oxidoreductases involved in such mechanisms can be considered as attractive molecular targets for the development of new therapeutic strategies. On the other hand, for more than two decades, transition metals derivatives have been leading the research on drugs as alternatives to platinum-based treatments. The success of such compounds is particularly due to their attractive redox kinetics properties, favorable oxidation states, as well as routes of action different to interactions with DNA, in which redox interactions are crucial. For instance, the activity of oxidoreductases such as PHD2 (prolyl hydroxylase domain-containing protein) which can regulate angiogenesis in tumors, LDH (lactate dehydrogenase) related to glycolysis, and enzymes, such as catalases, SOD (superoxide dismutase), TRX (thioredoxin) or GSH (glutathione) involved in controlling oxidative stress, can be altered by metal effectors. In this review, we wish to discuss recent results on how transition metal complexes have been rationally designed to impact on redox processes, in search for effective and more specific cancer treatments.
Collapse
Affiliation(s)
- María Isabel Murillo
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Christian Gaiddon
- Strasbourg Université, Inserm UMR_S U1113, IRFAC, Strasbourg, France
| | - Ronan Le Lagadec
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
- *Correspondence: Ronan Le Lagadec,
| |
Collapse
|
11
|
Singlet Oxygen, Photodynamic Therapy, and Mechanisms of Cancer Cell Death. JOURNAL OF ONCOLOGY 2022; 2022:7211485. [PMID: 35794980 PMCID: PMC9252714 DOI: 10.1155/2022/7211485] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 01/06/2023]
Abstract
Photodynamic therapy (PDT) can be developed into an important arsenal against cancer; it is a minimally invasive therapy, which is used in the treatment or/and palliation of a variety of cancers and benign diseases. The removal of cancerous tissue is achieved with the use of photosensitizer and a light source, which excites the photosensitizer. This excitation causes the photosensitizer to generate singlet oxygen and other reactive oxygen species. PDT has been used in several types of cancers including nonmelanoma skin cancer, bladder cancer, esophageal cancer, head and neck cancer, and non-small cell lung cancer (NSCLC). Although it is routinely used in nonmelanoma skin cancer, it has not been widely adopted in other solid cancers due to a lack of clinical data showing the superiority of PDT over other forms of treatment. Singlet oxygen used in PDT can alter the activity of the catalase, which induces immunomodulation through HOCl signaling. The singlet oxygen can induce apoptosis through both the extrinsic and intrinsic pathways. The extrinsic pathway of apoptosis starts with the activation of the Fas receptor by singlet oxygen that leads to activation of the caspase-7 and caspase-3. In the case of the intrinsic pathway, disruption caused by singlet oxygen in the mitochondria membrane leads to the release of cytochrome c, which binds with APAF-1 and procaspase-9, forming a complex, which activates caspase-3. Mechanisms of PDT action can vary according to organelles affected. In the plasma membrane, membrane disruption is caused by the oxidative stress leading to the intake of calcium ions, which causes swelling and rupture of cells due to excess intake of water, whereas disruption of lysosome causes the release of the cathepsins B and D, which cleave Bid into tBid, which changes the mitochondrial outer membrane permeability (MOMP). Oxidative stress causes misfolding of protein in the endoplasmic reticulum. When misfolding exceeds the threshold, it triggers unfolding protein response (UPR), which leads to activation of caspase-9 and caspase-3. Finally, the activation of p38 MAPK works as an alternative pathway for the induction of MOMP.
Collapse
|
12
|
Gouveia-Fernandes S, Rodrigues A, Nunes C, Charneira C, Nunes J, Serpa J, Antunes AMM. Glycidamide and cis-2-butene-1,4-dial (BDA) as potential carcinogens and promoters of liver cancer - An in vitro study. Food Chem Toxicol 2022; 166:113251. [PMID: 35750087 DOI: 10.1016/j.fct.2022.113251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/15/2022] [Accepted: 06/18/2022] [Indexed: 10/18/2022]
Abstract
Acrylamide and furan are environmental and food contaminants that are metabolized by cytochrome P450 2E1 (CYP2E1), giving rise to glycidamide and cis-2-butene-1,4-dial (BDA) metabolites, respectively. Both glycidamide and BDA are electrophilic species that react with nucleophilic groups, being able to introduce mutations in DNA and perform epigenetic remodeling. However, whereas these carcinogens are primarily metabolized in the liver, the carcinogenic potential of acrylamide and furan in this organ is still controversial, based on findings from experimental animal studies. With the ultimate goal of providing further insights into this issue, we explored in vitro, using a hepatocyte cell line and a hepatocellular carcinoma cell line, the putative effect of these metabolites as carcinogens and cancer promoters. Molecular alterations were investigated in cells that survive glycidamide and BDA toxicity. We observed that those cells express CD133 stemness marker, present a high proliferative capacity and display an adjusted expression profile of genes encoding enzymes involved in oxidative stress control, such as GCL-C, GSTP1, GSTA3 and CAT. These molecular changes seem to be underlined, at least in part, by epigenetic remodeling involving histone deacetylases (HDACs). Although more studies are needed, here we present more insights towards the carcinogenic capacity of glycidamide and BDA and also point out their effect in favoring hepatocellular carcinoma progression.
Collapse
Affiliation(s)
- Sofia Gouveia-Fernandes
- NOVA Medical School Research, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal
| | - Armanda Rodrigues
- NOVA Medical School Research, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal
| | - Carolina Nunes
- NOVA Medical School Research, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal
| | - Catarina Charneira
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico, Av. Rovisco Pais, 1049 001, Lisboa, Portugal
| | - João Nunes
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico, Av. Rovisco Pais, 1049 001, Lisboa, Portugal
| | - Jacinta Serpa
- NOVA Medical School Research, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal.
| | - Alexandra M M Antunes
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico, Av. Rovisco Pais, 1049 001, Lisboa, Portugal.
| |
Collapse
|
13
|
The Effect of Encapsulated Apigenin Nanoparticles on HePG-2 Cells through Regulation of P53. Pharmaceutics 2022; 14:pharmaceutics14061160. [PMID: 35745733 PMCID: PMC9228521 DOI: 10.3390/pharmaceutics14061160] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/19/2022] [Accepted: 05/26/2022] [Indexed: 01/27/2023] Open
Abstract
Apigenin (Ap) is one of the most important natural flavonoids that has potent anticancer activity. This study was designed, for the first time, to load Ap into chitosan to improve its hydrophobicity and then it was coated with albumin-folic acid to increase its stability and bioavailability and to target cancer cells. The newly developed encapsulated Ap (Ap-CH-BSA-FANPs) was characterized and tested in vitro. The zeta potential of −17.0 mV was within the recommended range (−30 mV to +30 mV), indicating that encapsulated apigenin would not quickly settle and would be suspended. The in vitro results proved the great anticancer activity of the encapsulated apigenin on HePG-2 cells compared to pure Ap. The treated HePG-2 cells with Ap-CH-BSA-FANPs demonstrated the induction of apoptosis by increasing p53 gene expression, arresting the cell cycle, increasing caspase-9 levels, and decreasing both the MMP9 gene and Bcl-2 protein expression levels. Moreover, the higher antioxidant activity of the encapsulated apigenin treatment was evident through increasing SOD levels and decreasing the CAT concentration. In conclusion, the Ap-CH-BSA-FANPs were easy to produce with low coast, continued drug release, good loading capacity, high solubility in physiological pH, and were more stable than the formerly Ap-loaded liposomes or PLGA. Moreover, Ap-CH-BSA-FANPs may be a promising chemotherapeutic agent in the treatment of HCC.
Collapse
|
14
|
Nath P, Maiti D. A review of the mutagenic potential of N-ethyl-N-nitrosourea (ENU) to induce hematological malignancies. J Biochem Mol Toxicol 2022; 36:e23067. [PMID: 35393684 DOI: 10.1002/jbt.23067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/05/2021] [Accepted: 03/23/2022] [Indexed: 12/12/2022]
Abstract
This review is intended to summarize the existing literature on the mutagenicity of N-ethyl-N-nitrosourea (ENU) in inducing hematological malignancies, including acute myeloid leukemia (AML) in mice. Blood or hematological malignancies are the most common malignant disorders seen in people of all age groups. Driven by a number of genetic alterations, leukemia rule out the normal proliferation and differentiation of hematopoietic stem cells (HSCs) and their progenitors in the bone marrow (BM) and severely affects blood functions. Out of all hematological malignancies, AML is the most aggressive type, with a high incidence and mortality rate. AML is found as either de novo or secondary therapeutic AML (t-AML). t-AML is a serious adverse consequence of alkylator chemotherapy to the cancer patient and alone constitutes about 10%-20% of all reported AML cases. Cancer patients who received alkylator chemotherapy are at an elevated risk of developing t-AML. ENU has a long history of use as a potent carcinogen that induces blood malignancies in mice and rats that are pathologically similar to human AML and t-AML. ENU, once entered into the body, circulates all over the body tissues and reaches BM. It creates an overall state of suppression within the BM by damaging the marrow cells, alkylating the DNA, and forming DNA adducts within the early and late hematopoietic stem and progenitor cells. The BM holds a weak DNA repair mechanism due to low alkyltransferase, and poly [ADP-ribose] polymerase (PARP) enzyme content often fails to obliterate those adducts, acting as a catalyst to bring genetic abnormalities, including point gene mutations as well as chromosomal alterations, for example, translocation and inversion. Taking advantage of ENU-induced immune-suppressed state and weak immune surveillance, these mutations remain viable and slowly give rise to transformed HSCs. This review also highlights the carcinogenic nature of ENU and the complex relation between the ENU's overall toxicity in the induction of hematological malignancies.
Collapse
Affiliation(s)
- Priyatosh Nath
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Agartala, Tripura, India
| | - Debasish Maiti
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Agartala, Tripura, India
| |
Collapse
|
15
|
Cai P, Sheng G, Jiang S, Wang D, Zhao Z, Huang M, Jin J. Comparative Proteomics Analysis Reveals the Reversal Effect of Cryptotanshinone on Gefitinib-Resistant Cells in Epidermal Growth Factor Receptor-Mutant Lung Cancer. Front Pharmacol 2022; 13:837055. [PMID: 35370706 PMCID: PMC8965640 DOI: 10.3389/fphar.2022.837055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/18/2022] [Indexed: 12/23/2022] Open
Abstract
Cryptotanshinone (CTS) is a lipophilic constituent of Salvia miltiorrhiza, with a broad-spectrum anticancer activity. We have observed that CTS enhances the efficacy of gefitinib in human lung cancer H1975 cells, yet little is known about its molecular mechanism. To explore how CTS enhances H1975 cell sensitivity to gefitinib, we figured out differential proteins of H1975 cells treated by gefitinib alone or in combination with CTS using label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein–protein interaction (PPI) bioinformatic analyses of the differential proteins were performed. CTS enhanced H1975 cell sensitivity to gefitinib in vitro and in vivo, with 115 and 128 differential proteins identified, respectively. GO enrichment, KEGG analysis, and PPI network comprehensively demonstrated that CTS mainly impacted the redox process and fatty acid metabolism in H1975 cells. Moreover, three differential proteins, namely, catalase (CAT), heme oxygenase 1 (HMOX1), and stearoyl-CoA desaturase (SCD) were validated by RT-qPCR and Western blot. In conclusion, we used a proteomic method to study the mechanism of CTS enhancing gefitinib sensitivity in H1975 cells. Our finding reveals the potential protein targets of CTS in overcoming gefitinib resistance, which may be therapeutical targets in lung cancer.
Collapse
Affiliation(s)
- Peiheng Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Gaofan Sheng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Shiqin Jiang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Daifei Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhongxiang Zhao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jing Jin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Jing Jin,
| |
Collapse
|
16
|
Abstract
Significance: Vitamin C (ascorbate), in regard to its effectiveness against malignancies, has had a controversial history in cancer treatment. It has been shown that in vitro and in vivo anticancer efficacy of ascorbate relies on its pro-oxidant effect mainly from an increased generation of reactive oxygen species (ROS). A growing understanding of its anticancer activities and pharmacokinetic properties has prompted scientists to re-evaluate the significance of ascorbate in cancer treatment. Recent Advances: A recent resurge in ascorbate research emerged after discovering that, at high doses, ascorbate preferentially kills Kirsten-Ras (K-ras)- and B-raf oncogene (BRAF)-mutant cancer cells. In addition, some of the main hallmarks of cancer cells, such as redox homeostasis and oxygen-sensing regulation (through inhibition of hypoxia-inducible factor-1 alpha [HIF-1α] activity), are affected by vitamin C. Critical Issues: Currently, there is no clear consensus from the literature in regard to the beneficial effects of antioxidants. Results from both human and animal studies provide no clear evidence about the benefit of antioxidant treatment in preventing or suppressing cancer development. Since pro-oxidants may affect both normal and tumor cells, the extremely low toxicity of ascorbate represents a main advantage. This guarantees the safe inclusion of ascorbate in clinical protocols to treat cancer patients. Future Directions: Current research could focus on elucidating the wide array of reactions between ascorbate and reactive species, namely ROS, reactive nitrogen species as well as reactive sulfide species, and their intracellular molecular targets. Unraveling these mechanisms could allow researchers to assess what could be the optimal combination of ascorbate with standard treatments.
Collapse
Affiliation(s)
- Christophe Glorieux
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, P. R. China
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile.,Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
17
|
Galasso M, Gambino S, Romanelli MG, Donadelli M, Scupoli MT. Browsing the oldest antioxidant enzyme: catalase and its multiple regulation in cancer. Free Radic Biol Med 2021; 172:264-272. [PMID: 34129927 DOI: 10.1016/j.freeradbiomed.2021.06.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/31/2021] [Accepted: 06/11/2021] [Indexed: 01/17/2023]
Abstract
Aerobic organisms possess numerous antioxidant enzymatic families, including catalases, superoxide dismutases (SODs), peroxiredoxins (PRDXs), and glutathione peroxidases (GPXs), which work cooperatively to protect cells from an excess of reactive oxygen species (ROS) derived from endogenous metabolism or external microenvironment. Catalase, as well as other antioxidant enzymes, plays an important dichotomous role in cancer. Therefore, therapies aimed at either reverting the increased or further escalating catalase levels could be effective, depending on the metabolic landscape and on the redox status of cancer cells. This dichotomous role of catalase in cancers highlights the importance to deepen comprehensively the role and the regulation of this crucial antioxidant enzyme. The present review highlights the role of catalase in cancer and provides a comprehensive description of the molecular mechanisms associated with the multiple levels of catalase regulation.
Collapse
Affiliation(s)
- Marilisa Galasso
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; Department of Medicine, University of Verona, Verona, Italy
| | - Simona Gambino
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Maria Grazia Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; Research Center LURM -Interdepartmental Laboratory of Medical Research, University of Verona, Verona, Italy.
| |
Collapse
|
18
|
Meyer MT, Watermann C, Dreyer T, Wagner S, Wittekindt C, Klussmann JP, Ergün S, Baumgart-Vogt E, Karnati S. Differential Expression of Peroxisomal Proteins in Distinct Types of Parotid Gland Tumors. Int J Mol Sci 2021; 22:7872. [PMID: 34360635 PMCID: PMC8345988 DOI: 10.3390/ijms22157872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
Salivary gland cancers are rare but aggressive tumors that have poor prognosis and lack effective cure. Of those, parotid tumors constitute the majority. Functioning as metabolic machinery contributing to cellular redox balance, peroxisomes have emerged as crucial players in tumorigenesis. Studies on murine and human cells have examined the role of peroxisomes in carcinogenesis with conflicting results. These studies either examined the consequences of altered peroxisomal proliferators or compared their expression in healthy and neoplastic tissues. None, however, examined such differences exclusively in human parotid tissue or extended comparison to peroxisomal proteins and their associated gene expressions. Therefore, we examined differences in peroxisomal dynamics in parotid tumors of different morphologies. Using immunofluorescence and quantitative PCR, we compared the expression levels of key peroxisomal enzymes and proliferators in healthy and neoplastic parotid tissue samples. Three parotid tumor subtypes were examined: pleomorphic adenoma, mucoepidermoid carcinoma and acinic cell carcinoma. We observed higher expression of peroxisomal matrix proteins in neoplastic samples with exceptional down regulation of certain enzymes; however, the degree of expression varied between tumor subtypes. Our findings confirm previous experimental results on other organ tissues and suggest peroxisomes as possible therapeutic targets or markers in all or certain subtypes of parotid neoplasms.
Collapse
Affiliation(s)
- Malin Tordis Meyer
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
| | - Christoph Watermann
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
| | - Thomas Dreyer
- Institute of Pathology, Justus Liebig University, Langhansstrasse 10, D-35392 Gießen, Germany;
| | - Steffen Wagner
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
| | - Claus Wittekindt
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
| | - Jens Peter Klussmann
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, D-50931 Cologne, Germany
| | - Süleyman Ergün
- Institute for Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Koellikerstrasse 6, D-97070 Würzburg, Germany;
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology II, Medical Cell Biology, Justus Liebig University, D-35385 Gießen, Germany;
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Koellikerstrasse 6, D-97070 Würzburg, Germany;
- Institute for Anatomy and Cell Biology II, Medical Cell Biology, Justus Liebig University, D-35385 Gießen, Germany;
| |
Collapse
|
19
|
Alsherbiny MA, Bhuyan DJ, Radwan I, Chang D, Li CG. Metabolomic Identification of Anticancer Metabolites of Australian Propolis and Proteomic Elucidation of Its Synergistic Mechanisms with Doxorubicin in the MCF7 Cells. Int J Mol Sci 2021; 22:ijms22157840. [PMID: 34360606 PMCID: PMC8346082 DOI: 10.3390/ijms22157840] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022] Open
Abstract
The combination of natural products with standard chemotherapeutic agents offers a promising strategy to enhance the efficacy or reduce the side effects of standard chemotherapy. Doxorubicin (DOX), a standard drug for breast cancer, has several disadvantages, including severe side effects and the development of drug resistance. Recently, we reported the potential bioactive markers of Australian propolis extract (AP-1) and their broad spectrum of pharmacological activities. In the present study, we explored the synergistic interactions between AP-1 and DOX in the MCF7 breast adenocarcinoma cells using different synergy quantitation models. Biochemometric and metabolomics-driven analysis was performed to identify the potential anticancer metabolites in AP-1. The molecular mechanisms of synergy were studied by analysing the apoptotic profile via flow cytometry, apoptotic proteome array and measuring the oxidative status of the MCF7 cells treated with the most synergistic combination. Furthermore, label-free quantification proteomics analysis was performed to decipher the underlying synergistic mechanisms. Five prenylated stilbenes were identified as the key metabolites in the most active AP-1 fraction. Strong synergy was observed when AP-1 was combined with DOX in the ratio of 100:0.29 (w/w) as validated by different synergy quantitation models implemented. AP-1 significantly enhanced the inhibitory effect of DOX against MCF7 cell proliferation in a dose-dependent manner with significant inhibition of the reactive oxygen species (p < 0.0001) compared to DOX alone. AP-1 enabled the reversal of DOX-mediated necrosis to programmed cell death, which may be advantageous to decline DOX-related side effects. AP-1 also significantly enhanced the apoptotic effect of DOX after 24 h of treatment with significant upregulation of catalase, HTRA2/Omi, FADD together with DR5 and DR4 TRAIL-mediated apoptosis (p < 0.05), contributing to the antiproliferative activity of AP-1. Significant upregulation of pro-apoptotic p27, PON2 and catalase with downregulated anti-apoptotic XIAP, HSP60 and HIF-1α, and increased antioxidant proteins (catalase and PON2) may be associated with the improved apoptosis and oxidative status of the synergistic combination-treated MCF7 cells compared to the mono treatments. Shotgun proteomics identified 21 significantly dysregulated proteins in the synergistic combination-treated cells versus the mono treatments. These proteins were involved in the TP53/ATM-regulated non-homologous end-joining pathway and double-strand breaks repairs, recruiting the overexpressed BRCA1 and suppressed RIF1 encoded proteins. The overexpression of UPF2 was noticed in the synergistic combination treatment, which could assist in overcoming doxorubicin resistance-associated long non-coding RNA and metastasis of the MCF7 cells. In conclusion, we identified the significant synergy and highlighted the key molecular pathways in the interaction between AP-1 and DOX in the MCF7 cells together with the AP-1 anticancer metabolites. Further in vivo and clinical studies are warranted on this synergistic combination.
Collapse
Affiliation(s)
- Muhammad A. Alsherbiny
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| | - Deep J. Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| | - Ibrahim Radwan
- Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia;
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
| | - Chun-Guang Li
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| |
Collapse
|
20
|
Lan W, Chen S, Nong G. An Efficient Synthesis of Novel Dextran-Arsenite Nanoparticles intended for Potential Antitumor Drug Material. AN ACAD BRAS CIENC 2021; 93:e20190551. [PMID: 33729378 DOI: 10.1590/0001-3765202120190551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/05/2019] [Indexed: 11/22/2022] Open
Abstract
The functionalization of polysaccharides with synthetic nanopolymers has attracted great attention owing to the applications of this method in many industrial fields. This work aimed to investigate the effect of arsenic trioxide on the functionalization of dextran. Dextran-arsenite nanoparticle formation was induced by microwave with sulfuric acid as a catalyst. Various analytical techniques were used to verify the structure of the nanopolymers. Besides, various reaction conditions, such as dextran concentration, arsenic trioxide concentration and pH, were investigated to determine their impact on particle size. The results indicated that the product was an arsenite-based nanomaterial retaining the basic configuration of dextran and that the product size was positively correlated with pH but negatively correlated with arsenic trioxide concentration. Moreover, the inhibitory effects of the dextran-arsenite nanoparticles on the growth of the human colorectal cancer cell line HCT-116 and human hepatoma carcinoma cell lines Huh-7 and SMMC-7721 were studied. The results showed that the product could inhibit the proliferation of these three tumor cell lines in a dose-dependent manner. Therefore, the product could be a new type of functional nanomaterial for further study on the synthesis, biological activity and development of polysaccharide drugs.
Collapse
Affiliation(s)
- Weibing Lan
- Guangxi University, College of Light Industry and Food Engineering, No.100, Daxue East Road, Nanning, 530004, Guangxi, China.,Beibu Gulf University, College of Food Engineering, No.12, Binhai Avenue, Qinzhou 535011, China
| | - Shan Chen
- Guangxi University, College of Light Industry and Food Engineering, No.100, Daxue East Road, Nanning, 530004, Guangxi, China
| | - Guangzai Nong
- Guangxi University, College of Light Industry and Food Engineering, No.100, Daxue East Road, Nanning, 530004, Guangxi, China
| |
Collapse
|
21
|
Yousefnia S. Mechanistic effects of arsenic trioxide on acute promyelocytic leukemia and other types of leukemias. Cell Biol Int 2021; 45:1148-1157. [PMID: 33527587 DOI: 10.1002/cbin.11563] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/25/2020] [Accepted: 01/31/2021] [Indexed: 12/13/2022]
Abstract
Acute promyelocytic leukemia (APL), a subtype of acute myeloid leukemia characterized with a translocation between promyelocytic leukemia gene (PML) on chromosome 15 and retinoic acid receptor alpha gene (RARα) on chromosome 17. Transcription of this fusion gene results in PML/RARα fusion protein blocking expression of critical genes involved in differentiation of myeloid cells through interaction with RAR element. PML/RARα fusion protein prevents normal function of PML and RARα as well as inhibiting apoptosis. Arsenic trioxide (ATO) is an important agent for the treatment of relapsed and newly diagnosed APL. ATO induces apoptosis, autophagy, and partial cellular differentiation as well as inhibiting cell growth and angiogenesis. Recognition of signaling pathways and molecular mechanisms induced by ATO can be effective for discovering novel treatment strategies to target leukemia cells. Also, it can be developed for the treatment of a variety of cancer cells. This review provides a perspective on anticancerous effects of ATO on APL and leukemia cells.
Collapse
Affiliation(s)
- Saghar Yousefnia
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
22
|
El-Mahdy MA, Alzarie YA, Hemann C, Badary OA, Nofal S, Zweier JL. The novel SOD mimetic GC4419 increases cancer cell killing with sensitization to ionizing radiation while protecting normal cells. Free Radic Biol Med 2020; 160:630-642. [PMID: 32739595 PMCID: PMC7704930 DOI: 10.1016/j.freeradbiomed.2020.07.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/23/2020] [Accepted: 07/25/2020] [Indexed: 02/08/2023]
Abstract
While radiotherapy is a widely used treatment for many types of human cancer, problems of radio-resistance and side effects remain. Side effects induced by ionizing radiation (IR) arise primarily from its propensity to trigger inflammation and oxidative stress with damage of normal cells and tissues near the treatment area. The highly potent superoxide dismutase mimetic, GC4419 (Galera Therapeutics), rapidly enters cells and is highly effective in dismutating superoxide (O2•-). We performed studies to assess the potency of GC4419 in cancer killing and radio-sensitization in human lung cancer cells and normal immortalized lung cells. Treatment with GC4419 did not alter the radical generation during IR, primarily hydroxyl radical (.OH); however, it quenched the increased levels of O2•- detected in the cancer cells before and following IR. GC4419 triggered cancer cell death and inhibited cancer cell proliferation with no adverse effect on normal cells. Combination of GC4419 with IR augmented the cytotoxic effects of IR on cancer cells compared to monotherapy, while protecting normal cells from IR-induced cell death. DNA fragmentation and caspase-3 activity assays showed that combination of GC4419 with IR enhances cancer cell apoptosis. Moreover, GC4419 increased IR-induced Bax levels with decreased Bcl-2 and elevated Bax/Bcl-2 ratio following treatment. GC4419 increased TrxR activity in the normal cells but decreased activity in cancer cells, conferring increased cancer cell sensitivity to oxidative stress. In conclusion, GC4419 increases the cytotoxic and pro-apoptotic activity of IR in lung cancer cells while decreasing injury in normal cells.
Collapse
Affiliation(s)
- Mohamed A El-Mahdy
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Yasmin A Alzarie
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA; National Organization of Drug Control and Research, Cairo, Egypt
| | - Craig Hemann
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Osama A Badary
- Department of Clinical Pharmacy, College of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Shahira Nofal
- Department of Pharmacology and Toxicology, College of Pharmacy, Helwan University, Cairo, Egypt
| | - Jay L Zweier
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
23
|
Selective Targeting of Cancerous Mitochondria and Suppression of Tumor Growth Using Redox-Active Treatment Adjuvant. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6212935. [PMID: 33204397 PMCID: PMC7652615 DOI: 10.1155/2020/6212935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/13/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
Redox-active substances and their combinations, such as of quinone/ascorbate and in particular menadione/ascorbate (M/A; also named Apatone®), attract attention with their unusual ability to kill cancer cells without affecting the viability of normal cells as well as with the synergistic anticancer effect of both molecules. So far, the primary mechanism of M/A-mediated anticancer effects has not been linked to the mitochondria. The aim of our study was to clarify whether this “combination drug” affects mitochondrial functionality specifically in cancer cells. Studies were conducted on cancer cells (Jurkat, Colon26, and MCF7) and normal cells (normal lymphocytes, FHC, and MCF10A), treated with different concentrations of menadione, ascorbate, and/or their combination (2/200, 3/300, 5/500, 10/1000, and 20/2000 μM/μM of M/A). M/A exhibited highly specific and synergistic suppression on cancer cell growth but without adversely affecting the viability of normal cells at pharmacologically attainable concentrations. In M/A-treated cancer cells, the cytostatic/cytotoxic effect is accompanied by (i) extremely high production of mitochondrial superoxide (up to 15-fold over the control level), (ii) a significant decrease of mitochondrial membrane potential, (iii) a decrease of the steady-state levels of ATP, succinate, NADH, and NAD+, and (iv) a decreased expression of programed cell death ligand 1 (PD-L1)—one of the major immune checkpoints. These effects were dose dependent. The inhibition of NQO1 by dicoumarol increased mitochondrial superoxide and sensitized cancer cells to M/A. In normal cells, M/A induced relatively low and dose-independent increase of mitochondrial superoxide and mild oxidative stress, which seems to be well tolerated. These data suggest that all anticancer effects of M/A result from a specific mechanism, tightly connected to the mitochondria of cancer cells. At low/tolerable doses of M/A (1/100-3/300 μM/μM) attainable in cancer by oral and parenteral administration, M/A sensitized cancer cells to conventional anticancer drugs, exhibiting synergistic or additive cytotoxicity accompanied by impressive induction of apoptosis. Combinations of M/A with 13 anticancer drugs were investigated (ABT-737, barasertib, bleomycin, BEZ-235, bortezomib, cisplatin, everolimus, lomustine, lonafarnib, MG-132, MLN-2238, palbociclib, and PI-103). Low/tolerable doses of M/A did not induce irreversible cytotoxicity in cancer cells but did cause irreversible metabolic changes, including: (i) a decrease of succinate and NADH, (ii) depolarization of the mitochondrial membrane, and (iii) overproduction of superoxide in the mitochondria of cancer cells only. In addition, M/A suppressed tumor growth in vivo after oral administration in mice with melanoma and the drug downregulated PD-L1 in melanoma cells. Experimental data suggest a great potential for beneficial anticancer effects of M/A through increasing the sensitivity of cancer cells to conventional anticancer therapy, as well as to the immune system, while sparing normal cells. We hypothesize that M/A-mediated anticancer effects are triggered by redox cycling of both substances, specifically within dysfunctional mitochondria. M/A may also have a beneficial effect on the immune system, making cancer cells “visible” and more vulnerable to the native immune response.
Collapse
|
24
|
Wang Y, Yang T, Han Y, Ren Z, Zou J, Liu J, Xi S. lncRNA OTUD6B-AS1 Exacerbates As 2O 3-Induced Oxidative Damage in Bladder Cancer via miR-6734-5p-Mediated Functional Inhibition of IDH2. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3035624. [PMID: 32952848 PMCID: PMC7481943 DOI: 10.1155/2020/3035624] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/06/2020] [Accepted: 07/18/2020] [Indexed: 12/18/2022]
Abstract
Arsenic trioxide (As2O3) is a promising effective chemotherapeutic agent for cancer treatment; however, how and through what molecular mechanisms the oxidative damage of As2O3 is controlled remains poorly understood. Recently, the involvement of dysregulated long noncoding RNA ovarian tumor domain containing 6B antisense RNA1 (lncRNA OTUD6B-AS1) in tumorigenesis is established. Here, for the first time, we characterize the regulation of As2O3 in the oxidative damage against bladder cancer via lncRNA OTUD6B-AS1. As2O3 could activate lncRNA OTUD6B-AS1 transcription in bladder cancer cells, and these findings were validated in a xenograft tumor model. Functional assays showed that lncRNA OTUD6B-AS1 dramatically exacerbated As2O3-mediated oxidative damage by inducing oxidative stress. Mechanistically, As2O3 increased levels of metal-regulatory transcription factor 1 (MTF1), which regulates lncRNA OTUD6B-AS1, in response to oxidative stress. Further, lncRNA OTUD6B-AS1 inhibited mitochondrial NADP+-dependent isocitrate dehydrogenase 2 (IDH2) expression by stabilizing miR-6734-5p, which contributed to cytotoxicity by enhancing oxidative stress. Together, our findings offer new insights into the mechanism of As2O3-induced oxidative damage and identify important factors in the pathway, As2O3/lncRNA OTUD6B-AS1/miR-6734-5p/IDH2, expanding the knowledge of activity of As2O3 as cancer treatment.
Collapse
Affiliation(s)
- Yutong Wang
- Department of Environmental Health, China Medical University, Shenyang 110122, China
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Tianyao Yang
- Department of Environmental Health, China Medical University, Shenyang 110122, China
| | - Yanshou Han
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Zhaozhou Ren
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jiayun Zou
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Jieyu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, China
| | - Shuhua Xi
- Department of Environmental Health, China Medical University, Shenyang 110122, China
| |
Collapse
|
25
|
Palma JM, Mateos RM, López-Jaramillo J, Rodríguez-Ruiz M, González-Gordo S, Lechuga-Sancho AM, Corpas FJ. Plant catalases as NO and H 2S targets. Redox Biol 2020; 34:101525. [PMID: 32505768 PMCID: PMC7276441 DOI: 10.1016/j.redox.2020.101525] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/20/2022] Open
Abstract
Catalase is a powerful antioxidant metalloenzyme located in peroxisomes which also plays a central role in signaling processes under physiological and adverse situations. Whereas animals contain a single catalase gene, in plants this enzyme is encoded by a multigene family providing multiple isoenzymes whose number varies depending on the species, and their expression is regulated according to their tissue/organ distribution and the environmental conditions. This enzyme can be modulated by reactive oxygen and nitrogen species (ROS/RNS) as well as by hydrogen sulfide (H2S). Catalase is the major protein undergoing Tyr-nitration [post-translational modification (PTM) promoted by RNS] during fruit ripening, but the enzyme from diverse sources is also susceptible to undergo other activity-modifying PTMs. Data on S-nitrosation and persulfidation of catalase from different plant origins are given and compared here with results from obese children where S-nitrosation of catalase occurs. The cysteine residues prone to be S-nitrosated in catalase from plants and from bovine liver have been identified. These evidences assign to peroxisomes a crucial statement in the signaling crossroads among relevant molecules (NO and H2S), since catalase is allocated in these organelles. This review depicts a scenario where the regulation of catalase through PTMs, especially S-nitrosation and persulfidation, is highlighted.
Collapse
Affiliation(s)
- José M Palma
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Dept. Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Granada, Spain.
| | - Rosa M Mateos
- Imflammation, Nutrition, Metabolism and Oxidative Stress Study Group (INMOX), Biomedical Research and Innovation Institute of Cádiz (INiBICA), Research Unit, Puerta del Mar University Hospital, Cádiz, Spain; Area of Biochemistry and Molecular Biology, Department of Biomedicine, Biotechnology and Public Health, University of Cádiz, Cádiz, Spain
| | | | - Marta Rodríguez-Ruiz
- Laboratório de Fisiologia do Desenvolvimiento Vegetal; Instituto de Biociências-Universidad de São Paulo; Cidade Universitária-São Paulo-SP, Brazil
| | - Salvador González-Gordo
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Dept. Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Granada, Spain
| | - Alfonso M Lechuga-Sancho
- Imflammation, Nutrition, Metabolism and Oxidative Stress Study Group (INMOX), Biomedical Research and Innovation Institute of Cádiz (INiBICA), Research Unit, Puerta del Mar University Hospital, Cádiz, Spain; Department of Child and Mother Health and Radiology, Medical School, University of Cádiz, Cádiz, Spain
| | - Francisco J Corpas
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Dept. Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Granada, Spain
| |
Collapse
|
26
|
Narayanan D, Ma S, Özcelik D. Targeting the Redox Landscape in Cancer Therapy. Cancers (Basel) 2020; 12:cancers12071706. [PMID: 32605023 PMCID: PMC7407119 DOI: 10.3390/cancers12071706] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are produced predominantly by the mitochondrial electron transport chain and by NADPH oxidases in peroxisomes and in the endoplasmic reticulum. The antioxidative defense counters overproduction of ROS with detoxifying enzymes and molecular scavengers, for instance, superoxide dismutase and glutathione, in order to restore redox homeostasis. Mutations in the redox landscape can induce carcinogenesis, whereas increased ROS production can perpetuate cancer development. Moreover, cancer cells can increase production of antioxidants, leading to resistance against chemo- or radiotherapy. Research has been developing pharmaceuticals to target the redox landscape in cancer. For instance, inhibition of key players in the redox landscape aims to modulate ROS production in order to prevent tumor development or to sensitize cancer cells in radiotherapy. Besides the redox landscape of a single cell, alternative strategies take aim at the multi-cellular level. Extracellular vesicles, such as exosomes, are crucial for the development of the hypoxic tumor microenvironment, and hence are explored as target and as drug delivery systems in cancer therapy. This review summarizes the current pharmaceutical and experimental interventions of the cancer redox landscape.
Collapse
Affiliation(s)
- Dilip Narayanan
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
| | - Sana Ma
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
| | - Dennis Özcelik
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
- current address: Chemistry | Biology | Pharmacy Information Center, ETH Zürich, Vladimir-Prelog-Weg 10, 8093 Zürich, Switzerland
- Correspondence:
| |
Collapse
|
27
|
Koim-Puchowska B, Drozdz-Afelt JM, Lamparski R, Menka A, Kaminski P. Antioxidant defence barrier of great tit Parus major nestlings in response to trace elements. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:20321-20334. [PMID: 32239412 PMCID: PMC7244610 DOI: 10.1007/s11356-020-08495-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 03/17/2020] [Indexed: 04/12/2023]
Abstract
Metals can have direct and indirect effects on the generation of reactive oxygen species in wild birds. The aim of this work has been to examine the effect of exposure to trace metals (copper Cu, iron Fe, cobalt Co, manganese Mn) on oxidative stress biomarkers such as lipoperoxidation TBARS and level of superoxide dismutase SOD, catalase CAT, and reduced glutathione GSH in the livers and kidneys of great tit Parus major nestlings (n = 165, 63 broods) living in polluted environments associated with soda plants and agricultural activities (Kujawy region) and from a reference site (Tuchola Forest), both in the north of Poland. As we predicted, the level of TBARS in both organs of chicks from polluted areas was higher than in those from reference site. This could be connected with Fe concentrations, particularly in areas adjacent to soda plants (livers Rs = 0.49, p < 0.002; kidneys Rs = 0.69, p < 0.001). We also showed differences in the level of antioxidants depending on the environment. CAT activity was higher in nestlings from Kujawy than in those from Tuchola. Meanwhile SOD activity (both organs) and GSH levels (kidneys) were lower in the polluted area compared to the reference site. Concentrations of Cu, Fe, Co, and Mn may play a role in regulating the antioxidant system components' activity.
Collapse
Affiliation(s)
- Beata Koim-Puchowska
- Department of Biotechnology, Kazimierz Wielki University, K.J. Poniatowski St12, 85-671, Bydgoszcz, Poland
| | - Joanna M. Drozdz-Afelt
- Department of Biotechnology, Kazimierz Wielki University, K.J. Poniatowski St12, 85-671, Bydgoszcz, Poland
| | - Robert Lamparski
- Department of Biology and Plant Protection, UTP University of Science and Technology, Prof. S. Kaliski St. 7, 85-796 Bydgoszcz, Poland
| | - Aleksandra Menka
- Department of Biotechnology, Kazimierz Wielki University, K.J. Poniatowski St12, 85-671, Bydgoszcz, Poland
| | - Piotr Kaminski
- Collegium Medicum in Bydgoszcz, Department of Ecology and Environmental Protection, Nicolaus Copernicus University, M. Skłodowska-Curie St. 9, 85-094 Bydgoszcz, Poland
- Faculty of Biological Sciences, Institute of Biotechnology and Environmental Protection, Department of Biotechnology, University of Zielona Góra, Prof. Szafran St. 1, 65-516 Zielona Góra, Poland
| |
Collapse
|
28
|
Zhang J, Yang Z, Zhang S, Xie Z, Han S, Wang L, Zhang B, Sun S. Investigation of endogenous malondialdehyde through fluorescent probe MDA-6 during oxidative stress. Anal Chim Acta 2020; 1116:9-15. [DOI: 10.1016/j.aca.2020.04.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/02/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022]
|
29
|
Sánchez-Virosta P, Espín S, Ruiz S, Panda B, Ilmonen P, Schultz SL, Karouna-Renier N, García-Fernández AJ, Eeva T. Arsenic-related oxidative stress in experimentally-dosed wild great tit nestlings. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 259:113813. [PMID: 31896481 DOI: 10.1016/j.envpol.2019.113813] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/13/2019] [Accepted: 12/13/2019] [Indexed: 05/24/2023]
Abstract
Arsenic (As) is broadly distributed due to natural and anthropogenic sources, and it may cause adverse effects in birds. However, research on other elements (Pb, Hg and Cd) has been prioritized, resulting in scarce data on As exposure and related effects in wild birds. One of the mechanisms responsible for As toxicity is oxidative stress. Therefore, the aim of this study was to investigate if environmentally relevant As levels affected oxidative stress biomarkers in great tits (Parus major). This is the first field experiment studying the effects of As on oxidative stress in wild passerines. Wild great tit nestlings were orally dosed with sodium arsenite (Control: water, Low dose: 0.2 μg g-1 d-1 and High dose: 1 μg g-1 d-1; from day 3 to day 13 post-hatching). We intended to reach As concentrations similar to those at which passerines are exposed to at actual polluted areas. We compared the responses to the experimental manipulations (High, Low and Control groups) with those in an As/metal-exposed population breeding close to a Cu-Ni smelter in Finland (Smelter group). A set of antioxidants (tGSH, GSH:GSSG ratio, CAT, SOD, GST and GPx), and oxidative damage biomarkers (lipid peroxidation, protein carbonylation, 8-hydroxy-2'-deoxyguanosine formation in DNA, and telomere length) were explored in blood. Arsenic administration had no significant effect on most of the biomarkers measured: only the CAT activity was lower in the High As group and the GPx activity was enhanced in the Smelter group compared to the Control. Our results suggest that the dose and duration of the As exposure was not enough to induce oxidative damage in red cells of great tit nestlings. In spite of this, nestlings dosed with 1 μg g-1 d-1 of sodium arsenite showed non-significantly higher oxidative stress biomarkers than controls, suggesting that we were close to an effect level for the redox-defense system. Oxidative effects at equivalent As levels combined with other stressors cannot be dismissed.
Collapse
Affiliation(s)
- Pablo Sánchez-Virosta
- Department of Biology, University of Turku, 20014 Turku, Finland; Toxicology and Risk Assessment Group, Department of Health Sciences, IMIB-Arrixaca, Faculty of Veterinary, University of Murcia, 30100 Murcia, Spain.
| | - Silvia Espín
- Department of Biology, University of Turku, 20014 Turku, Finland; Toxicology and Risk Assessment Group, Department of Health Sciences, IMIB-Arrixaca, Faculty of Veterinary, University of Murcia, 30100 Murcia, Spain.
| | - Sandra Ruiz
- Department of Biology, University of Turku, 20014 Turku, Finland
| | - Bineet Panda
- Department of Biology, University of Turku, 20014 Turku, Finland
| | - Petteri Ilmonen
- Department of Biology, University of Turku, 20014 Turku, Finland
| | - Sandra L Schultz
- U. S. Geological Survey, Patuxent Wildlife Research Center, Beltsville, MD 20705, USA
| | | | - Antonio J García-Fernández
- Toxicology and Risk Assessment Group, Department of Health Sciences, IMIB-Arrixaca, Faculty of Veterinary, University of Murcia, 30100 Murcia, Spain
| | - Tapio Eeva
- Department of Biology, University of Turku, 20014 Turku, Finland
| |
Collapse
|
30
|
Nazmeen A, Chen G, Ghosh TK, Maiti S. Breast cancer pathogenesis is linked to the intra-tumoral estrogen sulfotransferase (hSULT1E1) expressions regulated by cellular redox dependent Nrf-2/NF κβ interplay. Cancer Cell Int 2020; 20:70. [PMID: 32158360 PMCID: PMC7057506 DOI: 10.1186/s12935-020-1153-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Estrogen sulfotransferase catalyzes conjugation of sulfuryl-group to estradiol/estrone and regulates E2 availability/activity via estrogen-receptor or non-receptor mediated pathways. Sulfoconjugated estrogen fails to bind estrogen-receptor (ER). High estrogen is a known carcinogen in postmenopausal women. Reports reveal a potential redox-regulation of hSULT1E1/E2-signalling. Further, oxidatively-regulated nuclear-receptor-factor 2 (Nrf2) and NFκβ in relation to hSULT1E1/E2 could be therapeutic-target via cellular redox-modification. METHODS Here, oxidative stress-regulated SULT1E1-expression was analyzed in human breast carcinoma-tissues and in rat xenografted with human breast-tumor. Tumor and its surrounding tissues were obtained from the district-hospital. Intracellular redox-environment of tumors was screened with some in vitro studies. RT-PCR and western blotting was done for SULT1E1 expression. Immunohistochemistry was performed to analyze SULT1E1/Nrf2/NFκβ localization. Tissue-histoarchitecture/DNA-stability (comet assay) studies were done. RESULTS Oxidative-stress induces SULT1E1 via Nrf2/NFκβ cooperatively in tumor-pathogenesis to maintain the required proliferative-state under enriched E2-environment. Higher malondialdehyde/non-protein-soluble-thiol with increased superoxide-dismutase/glutathione-peroxidase/catalase activities was noticed. SULT1E1 expression and E2-level were increased in tumor-tissue compared to their corresponding surrounding-tissues. CONCLUSIONS It may be concluded that tumors maintain a sustainable oxidative-stress through impaired antioxidants as compared to the surrounding. Liver-tissues from xenografted rat manifested similar E2/antioxidant dysregulations favoring pre-tumorogenic environment.
Collapse
Affiliation(s)
- Aarifa Nazmeen
- Dept. of Biochemistry, Cell & Molecular Therapeutics Lab, Oriental Institute of Science & Technology, Midnapore, 721101 India
| | - Guangping Chen
- Venture I OSU Laboratory, Oklahoma Technology & Research Park, 1110 S. Innovation Way, Stillwater, OK 74074 USA
| | - Tamal Kanti Ghosh
- Special Secretary, Higher Medical Education, Health and Family Welfare Dept, Govt. of West Bengal, Salt Lake, Calcutta, India
| | - Smarajit Maiti
- Dept. of Biochemistry, Cell & Molecular Therapeutics Lab, Oriental Institute of Science & Technology, Midnapore, 721101 India
- Department of Biochemistry and Biotechnology, Cell & Molecular Therapeutics Lab, OIST, Midnapore, 721102 India
| |
Collapse
|
31
|
Chen H, Liu G, Qiao N, Kang Z, Hu L, Liao J, Yang F, Pang C, Liu B, Zeng Q, Li Y, Li Y. Toxic effects of arsenic trioxide on spermatogonia are associated with oxidative stress, mitochondrial dysfunction, autophagy and metabolomic alterations. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 190:110063. [PMID: 31846860 DOI: 10.1016/j.ecoenv.2019.110063] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/19/2019] [Accepted: 12/06/2019] [Indexed: 06/10/2023]
Abstract
Arsenic is a toxic metalloid that can cause male reproductive malfunctions and is widely distributed in the environment. The aim of this study was to investigate the cytotoxicity of arsenic trioxide (ATO) induced GC-1 spermatogonial (spg) cells. Our results found that ATO increased the levels of catalase (CAT) and malonaldehyde (MDA) and reactive oxygen species (ROS), while decreasing glutathione (GSH) and the total antioxidant capacity (T-AOC). Therefore, ATO triggered oxidative stress in GC-1 spg cells. In addition, ATO also caused severe mitochondrial dysfunction that included an increase in residual oxygen consumption (ROX), and decreased the routine respiration, maximal and ATP-linked respiration (ATP-L-R), as well as spare respiratory capacity (SRC), and respiratory control rate (RCR); ATO also damaged the mitochondrial structure, including mitochondrial cristae disordered and dissolved, mitochondrial vacuolar degeneration. Moreover, degradation of p62, LC3 conversion, increasing the number of acidic vesicle organelles (AVOs) and autophagosomes and autolysosomes are demonstrated that the cytotoxicity of ATO may be associated with autophagy. Meanwhile, the metabolomics analysis results showed that 20 metabolites (10 increased and 10 decreased) were significantly altered with the ATO exposure, suggesting that maybe there are the perturbations in amino acid metabolism, lipid metabolism, glycan biosynthesis and metabolism, metabolism of cofactors and vitamins. We concluded that ATO was toxic to GC-1 spg cells via inducing oxidative stress, mitochondrial dysfunction and autophagy as well as the disruption of normal metabolism. This study will aid our understanding of the mechanisms behind ATO-induced spermatogenic toxicity.
Collapse
Affiliation(s)
- Hanming Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| | - Gaoyang Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Na Qiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenlong Kang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Fan Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Congying Pang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Bingxian Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qiwen Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yao Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
32
|
Firczuk M, Bajor M, Graczyk-Jarzynka A, Fidyt K, Goral A, Zagozdzon R. Harnessing altered oxidative metabolism in cancer by augmented prooxidant therapy. Cancer Lett 2020; 471:1-11. [DOI: 10.1016/j.canlet.2019.11.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/20/2019] [Accepted: 11/30/2019] [Indexed: 12/17/2022]
|
33
|
Garufi A, Traversi G, Gilardini Montani MS, D'Orazi V, Pistritto G, Cirone M, D'Orazi G. Reduced chemotherapeutic sensitivity in high glucose condition: implication of antioxidant response. Oncotarget 2019; 10:4691-4702. [PMID: 31384396 PMCID: PMC6659798 DOI: 10.18632/oncotarget.27087] [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: 04/23/2019] [Accepted: 07/05/2019] [Indexed: 12/19/2022] Open
Abstract
Resistance to chemotherapy represents a major obstacle to successful treatment. The generation of reactive oxygen species (ROS) has been directly linked to the cytotoxic effects of several antitumor agents, including Adriamycin (ADR), and modulation of the oxidative balance has been implicated in the development and/or regulation of resistance to chemotherapeutic drugs. We recently showed that high glucose (HG) markedly diminished the cancer cell death induced by anticancer agents such as ADR. In the present study we attempted to evaluate the mechanism that impaired the cytotoxic effect of ADR in HG. We found that, in colon cancer cells, HG attenuated ADR-induced ROS production that consequently diminished ADR-induced H2AX phosphorylation and micronuclei (MN) formation. Mechanistically, HG attenuation of ADR-induced ROS production correlated with increased antioxidant response promoted by NRF2 activity. Thus, pharmacologic inhibition of NRF2 pathway by brusatol re-established the ADR cytotoxic effect impaired by HG. Together, the data provide new insights into chemotherapeutic-resistance mechanisms in HG condition dictated by increased NRF2-induced antioxidant response and how they may be overcome in order to restore chemosensitivity and ADR-induced cell death.
Collapse
Affiliation(s)
- Alessia Garufi
- IRCCS Regina Elena National Cancer Institute, Department of Research, Rome 00144, Italy.,University 'G. d'Annunzio', Department of Medical and Biotechnological Sciences, Chieti 66013, Italy
| | - Gianandrea Traversi
- IRCCS Regina Elena National Cancer Institute, Department of Research, Rome 00144, Italy.,University 'G. d'Annunzio', Department of Medical and Biotechnological Sciences, Chieti 66013, Italy
| | | | | | - Giuseppa Pistritto
- University Tor Vergata, Department of Systems Medicine, Rome 00133, Italy
| | - Mara Cirone
- Sapienza University, Department of Experimental Medicine, Rome 00161, Italy
| | - Gabriella D'Orazi
- IRCCS Regina Elena National Cancer Institute, Department of Research, Rome 00144, Italy.,University 'G. d'Annunzio', Department of Medical and Biotechnological Sciences, Chieti 66013, Italy
| |
Collapse
|
34
|
Abstract
Genomic instability is a common feature of tumours that has a wide range of disruptive effects on cellular homeostasis. In this review we briefly discuss how instability comes about, then focus on the impact of gain or loss of DNA (aneuploidy) on oxidative stress. We discuss several mechanisms that lead from aneuploidy to the production of reactive oxygen species, including the effects on protein complex stoichiometry, endoplasmic reticulum stress and metabolic disruption. Each of these are involved in positive feedback loops that amplify relatively minor genetic changes into major cellular disruption or cell death, depending on the capacity of the cell to induce antioxidants or processes such as mitophagy that can moderate the disruption. Finally we examine the direct effects of reactive oxygen species on mitosis and how oxidative stress can compromise centrosome number, cytoskeletal integrity and signalling processes that are vital for mitotic fidelity.
Collapse
Affiliation(s)
- David L Newman
- a Department of Molecular and Biomedical Science, University of Adelaide , Adelaide , Australia
| | - Lauren A Thurgood
- b Discipline of Molecular Medicine and Pathology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University , Adelaide , Australia
| | - Stephen L Gregory
- a Department of Molecular and Biomedical Science, University of Adelaide , Adelaide , Australia.,b Discipline of Molecular Medicine and Pathology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University , Adelaide , Australia
| |
Collapse
|
35
|
Mutant p53 and Cellular Stress Pathways: A Criminal Alliance That Promotes Cancer Progression. Cancers (Basel) 2019; 11:cancers11050614. [PMID: 31052524 PMCID: PMC6563084 DOI: 10.3390/cancers11050614] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 02/07/2023] Open
Abstract
The capability of cancer cells to manage stress induced by hypoxia, nutrient shortage, acidosis, redox imbalance, loss of calcium homeostasis and exposure to drugs is a key factor to ensure cancer survival and chemoresistance. Among the protective mechanisms utilized by cancer cells to cope with stress a pivotal role is played by the activation of heat shock proteins (HSP) response, anti-oxidant response induced by nuclear factor erythroid 2-related factor 2 (NRF2), the hypoxia-inducible factor-1 (HIF-1), the unfolded protein response (UPR) and autophagy, cellular processes strictly interconnected. However, depending on the type, intensity or duration of cellular stress, the balance between pro-survival and pro-death pathways may change, and cell survival may be shifted into cell death. Mutations of p53 (mutp53), occurring in more than 50% of human cancers, may confer oncogenic gain-of-function (GOF) to the protein, mainly due to its stabilization and interaction with the above reported cellular pathways that help cancer cells to adapt to stress. This review will focus on the interplay of mutp53 with HSPs, NRF2, UPR, and autophagy and discuss how the manipulation of these interconnected processes may tip the balance towards cell death or survival, particularly in response to therapies.
Collapse
|
36
|
Xu N, Yang Y, Chen L, Xu L, Xu X, Lin J. Enhancing the Effect of Pharmacological Ascorbate in Cancer Therapy via Acid‐Triggered Ferritin Nanoparticles. ACTA ACUST UNITED AC 2019; 3:e1900006. [DOI: 10.1002/adbi.201900006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/20/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Nuo Xu
- Synthetic and Functional Biomolecules CenterBeijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringInnovation Center for GenomicsPeking University Beijing 100871 China
| | - Yuan‐Fan Yang
- Synthetic and Functional Biomolecules CenterBeijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringInnovation Center for GenomicsPeking University Beijing 100871 China
| | - Long Chen
- Synthetic and Functional Biomolecules CenterBeijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringInnovation Center for GenomicsPeking University Beijing 100871 China
| | - Liang Xu
- Synthetic and Functional Biomolecules CenterBeijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringInnovation Center for GenomicsPeking University Beijing 100871 China
| | - Xiao‐Jie Xu
- Zhe Jiang Hisun Pharmaceutical Company Limited No.46 Waisha Road Taizhou Zhejiang China
| | - Jian Lin
- Synthetic and Functional Biomolecules CenterBeijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringInnovation Center for GenomicsPeking University Beijing 100871 China
| |
Collapse
|
37
|
Peroxisomes and cancer: The role of a metabolic specialist in a disease of aberrant metabolism. Biochim Biophys Acta Rev Cancer 2018; 1870:103-121. [PMID: 30012421 DOI: 10.1016/j.bbcan.2018.07.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/30/2018] [Accepted: 07/10/2018] [Indexed: 01/02/2023]
Abstract
Cancer is irrevocably linked to aberrant metabolic processes. While once considered a vestigial organelle, we now know that peroxisomes play a central role in the metabolism of reactive oxygen species, bile acids, ether phospholipids (e.g. plasmalogens), very-long chain, and branched-chain fatty acids. Immune system evasion is a hallmark of cancer, and peroxisomes have an emerging role in the regulation of cellular immune responses. Investigations of individual peroxisome proteins and metabolites support their pro-tumorigenic functions. However, a significant knowledge gap remains regarding how individual functions of proteins and metabolites of the peroxisome orchestrate its potential role as a pro-tumorigenic organelle. This review highlights new advances in our understanding of biogenesis, enzymatic functions, and autophagic degradation of peroxisomes (pexophagy), and provides evidence linking these activities to tumorigenesis. Finally, we propose avenues that may be exploited to target peroxisome-related processes as a mode of combatting cancer.
Collapse
|