1
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
2
|
Jaksic Karisik M, Jović Orsini N, Carkic J, Lazarevic M, Mitić D, Jokanovic B, Jokanović V, Milasin J. A Carbon-Based Nanomaterial with Dichotomous Effects: Antineoplastic on Oral Cancer Cells and Osteoinductive/Chondroinductive on Dental Pulp Stem Cells. J Funct Biomater 2025; 16:109. [PMID: 40137387 PMCID: PMC11943258 DOI: 10.3390/jfb16030109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Oral cancer is an aggressive malignancy with modest survival rates. It also causes disfigurement following surgical removal of the tumor, thus highlighting the need for new cancer treatment and tissue repair modalities. Carbon-based nanomaterials have emerged as promising tools in both anticancer and regenerative therapies. OBJECTIVES We aimed to synthesize a new carbon-based nanomaterial (CBN) and test its antineoplastic effects, as well as its potential regenerative capacity. MATERIALS AND METHODS A carbon nanomaterial, obtained by ball milling graphite flakes, was functionalized with polyvinylpyrrolidone (CBN/PVP). Its physicochemical properties were explored with X-ray diffraction (XRD), attenuated total reflection-Fourier transform infrared spectroscopy (ATR-FTIR), micro-Raman spectroscopy, fluorescent and scanning electron microscopy, and wettability analysis. For the antineoplastic effects investigation, oral cancer cells were treated with CBN/PVP and examined with MTT and migration assays, as well as cell-cycle and ROS production analyses. Gene expression was determined by qPCR. To examine the pro-regenerative capacity of CBN/PVP, dental pulp stem cell cultures (DPSCs) were treated with the nanomaterial and subjected to osteo- and chondro-induction. RESULTS Lower concentrations of CBN/PVP (50, 100 μg/mL) applied on cancer cells exerted remarkable cytotoxic effects, induced G1 cell-cycle arrest, and reduced cancer cell invasion potential by different mechanisms, including downregulation of the PI3K/AKT/mTOR pathway. In contrast, the addition of 50 µg/mL of CBN/PVP to DPSCs stimulated their survival and proliferation. CBN/PVP significantly enhanced both the osteogenic (p < 0.05) and chondrogenic (p < 0.01) induction of DPSCs. CONCLUSIONS The novel carbon-based nanomaterial displays unique characteristics, making it suitable in anticancer and regenerative therapies concomitantly.
Collapse
Affiliation(s)
- Milica Jaksic Karisik
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| | - Nataša Jović Orsini
- Vinča Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia;
| | - Jelena Carkic
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| | - Milos Lazarevic
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| | - Dijana Mitić
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| | | | | | - Jelena Milasin
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| |
Collapse
|
3
|
Jhetam Z, Martins-Furness C, Slabber C, Munro OQ, Nel M, Harmse L. Copper complexes induce haem oxygenase-1 (HMOX1) and cause apoptotic cell death in pancreatic cancer cells. J Inorg Biochem 2025; 264:112815. [PMID: 39740375 DOI: 10.1016/j.jinorgbio.2024.112815] [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: 07/27/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common pancreatic malignancy, has a dismal 5-year survival rate, making palliative chemotherapy the only treatment option. Targeted therapy has limited efficacy in PDAC, underscoring the need for novel therapeutic approaches. The inducible stress-response protein, haem oxygenase-1 (HMOX1), has been implicated in treatment failure in PDAC. Copper coordination complexes have shown promise as anticancer agents against various cancers, and are associated with apoptotic cell death. The different ligands to which copper is complexed, determine the specificity and efficacy of each complex. Three different classes of copper complexes were evaluated for anti-cancer activity against AsPC-1 and MIA PaCa-2 pancreatic cancer cell lines. A copper-phenanthroline-theophylline complex (CuPhTh2), a copper-8-aminoquinoline-naphthyl complex (Cu8AqN), and two copper-aromatic-isoindoline complexes (CuAIsI) were effective inhibitors of cell proliferation with clinically relevant IC50 values below 5 μM. The copper complexes caused reactive oxygen species (ROS) formation, promoted annexin-V binding, disrupted the mitochondrial membrane potential (MMP) and activated caspase-9 and caspase-3/7, confirming apoptotic cell death. Expression of nuclear HMOX1 was increased in both cell lines, with the CuPhTh2 complex being the most active. Inhibition of HMOX1 activity significantly decreased the IC50 values of these copper complexes suggesting that HMOX1 inhibition may alter treatment outcomes in PDAC.
Collapse
Affiliation(s)
- Zakeeya Jhetam
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Carla Martins-Furness
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Cathy Slabber
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg 2017, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg 2017, South Africa; School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK
| | - Marietha Nel
- Dept of Surgery, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Leonie Harmse
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa.
| |
Collapse
|
4
|
Penalva-Olcina R, Juan C, Fernández-Franzón M, Juan-García A. Neurotoxic implications of gliotoxin and ochratoxin A in SH-SY5Y cells: ROS-induced apoptosis and genotoxicity. Toxicol Lett 2025; 405:51-58. [PMID: 39947449 DOI: 10.1016/j.toxlet.2025.02.004] [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: 12/05/2024] [Revised: 01/20/2025] [Accepted: 02/11/2025] [Indexed: 02/16/2025]
Abstract
Gliotoxin (GTX) and ochratoxin A (OTA) are naturally produced toxins by fungi and are known for their potential health risks. With the aim of shed some light on the mechanisms by which GTX, OTA, and their combination exert toxicity at neuronal level, the following in vitro studies were conducted in SH-SY5Y cells: a) intracellular ROS monitorization by the H2-DCFDA assay b) study of the expression of pro-apoptotic genes Bcl2, Casp-3, and Bax by RT-qPCR c) study of the apoptotic-necrotic progression of SH-SY5Y cells by flow cytometry; d) study of the genotoxic potential through the in vitro micronucleus (MN) assay also by flow cytometry following OECD TG 487 guidelines. ROS production was increased when cells were exposed to mycotoxins at all scenarios tested highlighting the effects of GTX. Regarding gene expression, increases of Bax and Casp-3 genes at 1.3- and 3- folds respectively were observed when cells were exposed to GTX at 0.75 μM, with a more prominent increase after exposure to the binary combination [GTX + OTA] at [0.2 + 0.1] µM, increasing 3 and 5-folds more, respectively when compared to the control. MN formation increased a 30 % compared to control when exposed to GTX at 0.4 μM, 43 % for OTA at 0.8 μM, with the highest increase observed when cells were exposed to the combination [GTX + OTA] at [0.2 + 1.5] μM, obtaining a 65 % more MN formation. Based on the results obtained, we can conclude that for the proposed scenarios of exposure to GTX, OTA, and their combination, genotoxic effects together with oxidative effects at neuronal level in SH-SY5Y cell line, were found to play a key role in their mechanisms of toxic action.
Collapse
Affiliation(s)
- Raquel Penalva-Olcina
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent Andrés Estellés s/n, Burjassot, València 46100, Spain
| | - Cristina Juan
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent Andrés Estellés s/n, Burjassot, València 46100, Spain
| | - Mónica Fernández-Franzón
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent Andrés Estellés s/n, Burjassot, València 46100, Spain
| | - Ana Juan-García
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent Andrés Estellés s/n, Burjassot, València 46100, Spain.
| |
Collapse
|
5
|
Mashayekhi-Sardoo H, Hedayati-Moghadam M, Baghcheghi Y. Hippocampal Apoptosis: Molecular Mechanisms Triggered by Toxic Cannabinoid Exposure: A Narrative Review. Neurotoxicology 2025:S0161-813X(25)00019-1. [PMID: 40024347 DOI: 10.1016/j.neuro.2025.02.006] [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: 01/05/2025] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Hippocampal apoptosis is increasingly recognized as a significant consequence of toxic cannabinoid exposure, with profound implications for cognitive function and mental health. This narrative review comprehensively examines the molecular mechanisms underlying cannabinoid-induced apoptosis, focusing on the interplay of various bioactive compounds and their effects on neuronal integrity. We begin by discussing the key players in cannabinoid biology, followed by a synthesis of findings from animal and clinical studies that highlight the neurotoxic potential of cannabinoids. Central to our analysis are the roles of neuroinflammation and oxidative stress, which exacerbate neuronal damage and contribute to cell death. The activation of cannabinoid receptors, particularly CB1 and CB2, is scrutinized for its dual role in mediating neuroprotective and neurotoxic effects. We explore calcium dysregulation as a critical mechanism that leads to excitotoxicity, mitochondrial dysfunction, and the activation of pro-apoptotic pathways. Additionally, we address the inhibition of anti-apoptotic proteins, induction of endoplasmic reticulum (ER) stress, and disruption of neurotransmitter systems, all of which further facilitate apoptosis in hippocampal neurons. Alterations in neurotrophic factor levels are also examined, as they play a vital role in neuronal survival and plasticity. Ultimately, this review underscores the multifaceted nature of cannabinoid-induced hippocampal apoptosis and calls for further research to elucidate these complex interactions, aiming to inform clinical practices and public health policies regarding cannabinoid use. The findings presented herein highlight the urgent need for a nuanced understanding of the risks associated with cannabinoid exposure, particularly in vulnerable populations.
Collapse
Affiliation(s)
- Habibeh Mashayekhi-Sardoo
- Student Research Committee Jiroft University of Medical Sciences, Jiroft, Iran; Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran; School of Health, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Mahdiyeh Hedayati-Moghadam
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran; Student Research Committee Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Yousef Baghcheghi
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran; Student Research Committee Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
6
|
Perez-Serna AA, Guzman-Llorens D, Dos Santos RS, Marroqui L. Bcl-2 and Bcl-xL in Diabetes: Contributions to Endocrine Pancreas Viability and Function. Biomedicines 2025; 13:223. [PMID: 39857806 PMCID: PMC11760435 DOI: 10.3390/biomedicines13010223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Diabetes is a chronic metabolic disorder whose prevalence increases every year, affecting more than 530 million adults worldwide. Type 1 (T1D) and type 2 diabetes (T2D), the most common forms of diabetes, are characterized by the loss of functional pancreatic β-cells, mostly due to apoptosis. B-cell leukemia/lymphoma 2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-xL), two anti-apoptotic proteins belonging to the Bcl-2 family, are crucial for regulating the intrinsic pathway of apoptosis. However, over the years, they have been implicated in many other cellular processes, including intracellular Ca2+ homeostasis and the regulation of mitochondrial metabolism. Thus, understanding the biological processes in which these proteins are involved may be crucial to designing new therapeutic targets. This review summarizes the roles of Bcl-2 and Bcl-xL in apoptosis and metabolic homeostasis. It focuses on how the dysregulation of Bcl-2 and Bcl-xL affects pancreatic β-cell function and survival, and the consequences for diabetes development.
Collapse
Affiliation(s)
- Atenea A. Perez-Serna
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Guzman-Llorens
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
| | - Reinaldo S. Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l’Almazara 11, 03203 Elche, Alicante, Spain
| | - Laura Marroqui
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
7
|
Hussein Z, Michel HE, El-Naga RN, El-Demerdash E, Mantawy EM. Coenzyme Q10 ameliorates cyclophosphamide-induced chemobrain by repressing neuronal apoptosis and preserving hippocampal neurogenesis: Mechanistic roles of Wnt/ β-catenin signaling pathway. Neurotoxicology 2024; 105:21-33. [PMID: 39209270 DOI: 10.1016/j.neuro.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Deterioration in the neurocognitive function of cancer patients referred to as "Chemobrain" is a devastating obstacle associated with cyclophosphamide (CYP). CYP is an alkylating agent, clinically utilized as an efficient anticancer and immunosuppressant. Coenzyme Q10 (CoQ10) is a worthwhile micronutrient with diverse biological activities embracing antioxidant, anti-apoptotic, and neuroprotective effects. The current experiment was designed for investigating the neuroprotective capability of CoQ10 versus CYP-elicited chemobrain in rats besides elucidating the causal molecular mechanisms. Male Sprague Dawley rats received CoQ10 (10 mg/kg, orally, once daily, for 10 days) and/or a single dose of CYP (200 mg/kg i.p. on day 7). CoQ10 counteracted CYP-induced cognitive and motor dysfunction as demonstrated by the findings of neurobehavioral tests (passive avoidance, Y maze, locomotion, and rotarod tests). Histopathological analysis further affirmed the neuroprotective abilities of CoQ10. CoQ10 effectually diminished CYP-provoked oxidative injury by restoring the antioxidant activity of catalase (CAT) enzyme while reducing malondialdehyde (MDA) levels. Besides, CoQ10 efficiently repressed CYP-induced neuronal apoptosis by downregulating the expression of Bax and caspase-3 while upregulating the Bcl-2 expression. Moreover, CoQ10 hampered CYP-provoked upregulation in acetylcholinesterase (AChE) activity. Furthermore, CoQ10 considerably augmented hippocampal neurogenesis by elevating the expressions of brain-derived neurotrophic factor (BDNF) and Ki-67. These promising neuroprotective effects can be credited to upregulating Wnt/β-catenin pathway as evidenced by the elevated expressions of Wnt-3a, β-catenin, and Phoshpo-glycogen synthase kinase-3 β (p-GSK-3β). Collectively, these findings proved the neuroprotective capabilities of CoQ10 against CYP-induced chemobrain through combating oxidative injury, repressing intrinsic apoptosis, boosting neurogenesis, and eventually upregulating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Zeina Hussein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Preclinical and Translational Research Center, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Preclinical and Translational Research Center, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
8
|
Zhang W, Shen M, Chu P, Wang T, Ji J, Ning X, Yin S, Zhang K. Molecular characterization of CIRBP from Takifugu fasciatus and its potential roles in cold-induced liver damage. Int J Biol Macromol 2024; 281:136492. [PMID: 39393746 DOI: 10.1016/j.ijbiomac.2024.136492] [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: 07/06/2023] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
As a potent stressor, environmental cold stress induces severe mitochondrial dysfunction with the overproduction of reactive oxygen species (ROS) in fish, resulting in liver damage. However, the molecular mechanisms underlying the cold-induced liver damage remain unclear. In the present study, the cold-inducible RNA-binding protein (CIRBP) from Takifugu fasciatus was characterized, and its role in cold-induced oxidative stress damage was investigated. An acute liver injury model was constructed by exposing T. fasciatus individuals to temperatures of 25, 19, and 13 °C. Cold exposure markedly induced histomorphological liver injury and triggered endogenous apoptosis and NLRP3 inflammatory response. Cold treatment significantly increased CIRBP gene expression. A similar expression pattern was detected for thioredoxin (TRX), suggesting that these two proteins play a role in the establishment of cold adaptation. CIRBP binds directly to the 3'-UTR of TRX. Furthermore, in vivo experiment showed that, when CIRBP expression in T. fasciatus is knocked down, the time to loss equilibrium significantly shortened at 13 °C. Taken together, our study revealed that CIRBP is a critical protective factor against cold induced liver damage and that the CIRBP/TRX pathway could function as an underlying mechanism for cold adaptation in teleosts.
Collapse
Affiliation(s)
- Wenwen Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Minghao Shen
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Peng Chu
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Tao Wang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China
| | - Jie Ji
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China
| | - Xianhui Ning
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China
| | - Shaowu Yin
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China.
| | - Kai Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China.
| |
Collapse
|
9
|
An X, Yu W, Liu J, Tang D, Yang L, Chen X. Oxidative cell death in cancer: mechanisms and therapeutic opportunities. Cell Death Dis 2024; 15:556. [PMID: 39090114 PMCID: PMC11294602 DOI: 10.1038/s41419-024-06939-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen-containing molecules generated as natural byproducts during cellular processes, including metabolism. Under normal conditions, ROS play crucial roles in diverse cellular functions, including cell signaling and immune responses. However, a disturbance in the balance between ROS production and cellular antioxidant defenses can lead to an excessive ROS buildup, causing oxidative stress. This stress damages essential cellular components, including lipids, proteins, and DNA, potentially culminating in oxidative cell death. This form of cell death can take various forms, such as ferroptosis, apoptosis, necroptosis, pyroptosis, paraptosis, parthanatos, and oxeiptosis, each displaying distinct genetic, biochemical, and signaling characteristics. The investigation of oxidative cell death holds promise for the development of pharmacological agents that are used to prevent tumorigenesis or treat established cancer. Specifically, targeting key antioxidant proteins, such as SLC7A11, GCLC, GPX4, TXN, and TXNRD, represents an emerging approach for inducing oxidative cell death in cancer cells. This review provides a comprehensive summary of recent progress, opportunities, and challenges in targeting oxidative cell death for cancer therapy.
Collapse
Affiliation(s)
- Xiaoqin An
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Wenfeng Yu
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Jinbao Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Li Yang
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China.
| | - Xin Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
10
|
Chen Y, Luo X, Xu B, Bao X, Jia H, Yu B. Oxidative Stress-Mediated Programmed Cell Death: a Potential Therapy Target for Atherosclerosis. Cardiovasc Drugs Ther 2024; 38:819-832. [PMID: 36522550 DOI: 10.1007/s10557-022-07414-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/04/2022] [Indexed: 12/23/2022]
Abstract
Nowadays, as a type of orderly and active death determined by genes, programmed cell death (PCD), including apoptosis, pyroptosis, ferroptosis, and necroptosis, has attracted much attention owing to its participation in numerous chronic cardiovascular diseases, especially atherosclerosis (AS), a canonical chronic inflammatory disease featured by lipid metabolism disturbance. Abundant researches have reported that PCD under distinct internal conditions fulfills different roles of atherosclerotic pathological processes, including lipid core expansion, leukocyte adhesion, and infiltration. Noteworthy, emerging evidence recently has also suggested that oxidative stress (OS), an imbalance of antioxidants and oxygen free radicals, has the potential to mediate PCD occurrence via multiple ways, including oxidization and deubiquitination. Interestingly, more recently, several studies have proposed that the mediating mechanisms could effect on the atherosclerotic initiation and progression significantly from variable aspects, so it is of great clinical importance to clarify how OS-mediated PCD and AS interact. Herein, with the aim of summarizing potential and sufficient atherosclerotic therapy targets, we seek to provide extensive analysis of the specific regulatory mechanisms of PCD mediated by OS and their multifaceted effects on the entire pathological atherosclerotic progression.
Collapse
Affiliation(s)
- Yuwu Chen
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Xing Luo
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Biyi Xu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Xiaoyi Bao
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Haibo Jia
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China.
| | - Bo Yu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| |
Collapse
|
11
|
Iksen, Witayateeraporn W, Hardianti B, Pongrakhananon V. Comprehensive review of Bcl-2 family proteins in cancer apoptosis: Therapeutic strategies and promising updates of natural bioactive compounds and small molecules. Phytother Res 2024; 38:2249-2275. [PMID: 38415799 DOI: 10.1002/ptr.8157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/04/2024] [Accepted: 01/29/2024] [Indexed: 02/29/2024]
Abstract
Cancer has a considerably higher fatality rate than other diseases globally and is one of the most lethal and profoundly disruptive ailments. The increasing incidence of cancer among humans is one of the greatest challenges in the field of healthcare. A significant factor in the initiation and progression of tumorigenesis is the dysregulation of physiological processes governing cell death, which results in the survival of cancerous cells. B-cell lymphoma 2 (Bcl-2) family members play important roles in several cancer-related processes. Drug research and development have identified various promising natural compounds that demonstrate potent anticancer effects by specifically targeting Bcl-2 family proteins and their associated signaling pathways. This comprehensive review highlights the substantial roles of Bcl-2 family proteins in regulating apoptosis, including the intricate signaling pathways governing the activity of these proteins, the impact of reactive oxygen species, and the crucial involvement of proteasome degradation and the stress response. Furthermore, this review discusses advances in the exploration and potential therapeutic applications of natural compounds and small molecules targeting Bcl-2 family proteins and thus provides substantial scientific information and therapeutic strategies for cancer management.
Collapse
Affiliation(s)
- Iksen
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacy, Sekolah Tinggi Ilmu Kesehatan Senior Medan, Medan, Indonesia
| | - Wasita Witayateeraporn
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Besse Hardianti
- Laboratory of Pharmacology and Clinical Pharmacy, Faculty of Health Sciences, Almarisah Madani University, South Sulawesi, Indonesia
| | - Varisa Pongrakhananon
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Unit, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
12
|
Zhou H, Sun X, Dai Y, Wang X, Dai Z, Li X. 14-3-3-η interacts with BCL-2 to protect human endothelial progenitor cells from ox-LDL-triggered damage. Cell Biol Int 2024; 48:290-299. [PMID: 38100125 DOI: 10.1002/cbin.12105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/31/2023] [Accepted: 11/18/2023] [Indexed: 02/15/2024]
Abstract
Oxidized low-density lipoprotein (ox-LDL) causes dysfunction of endothelial progenitor cells (EPCs), and we recently reported that 14-3-3-η can attenuate the damage triggered by ox-LDL in EPCs. However, the molecular mechanisms by which 14-3-3-η protects EPCs from the damage caused by ox-LDL are not fully understood. In this study, we observed that the expression of 14-3-3-η and BCL-2 were downregulated in ox-LDL-treated EPCs. Overexpression of 14-3-3-η in ox-LDL-treated EPC significantly increased BCL-2 level, while knockdown of BCL-2 reduced 14-3-3-η expression and mitigated the protective effect of 14-3-3-η on EPCs. In addition, we discovered that 14-3-3-η colocalizes and interacts with BCL-2 in EPCs. Taken together, these data suggest that 14-3-3-η protects EPCs from ox-LDL-induced damage by its interaction with BCL-2.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaopei Sun
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi Dai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaotong Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhihong Dai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiuli Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
13
|
Shaheer K, Prabhu BS, Ali HS, Lakshmanan-M D. Breast cancer cells are sensitized by piperine to radiotherapy through estrogen receptor-α mediated modulation of a key NHEJ repair protein- DNA-PK. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155126. [PMID: 37913642 DOI: 10.1016/j.phymed.2023.155126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/03/2023] [Accepted: 09/27/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Non-homologous end joining, an important DNA-double-stranded break repair pathway, plays a prominent role in conferring resistance to radiotherapeutic agents, resulting in cancer progression and relapse. PURPOSE The molecular players involved in the radio-sensitizing effects of piperine and many other phytocompounds remain evasive to a great extent. The study is designed to assess if piperine, a plant alkaloid can alter the radioresistance by modulating the expression of non-homologous end-joining machinery. METHODS AND MATERIALS Estrogen receptor-positive/negative, breast cancer cells were cultured to understand the synergetic effects of piperine with radiotherapy. Cisplatin and Bazedoxifene were used as positive controls. Cells were exposed to γ- radiation using Low Dose gamma Irradiator-2000. The piperine effect on Estrogen receptor modulation, DNA-Damage, DNA-Damage-Response, and apoptosis was done by western blotting, immunofluorescence, yeast-based-estrogen-receptor-LacZ-reporter assay, and nuclear translocation analysis. Micronuclei assay was done for DNA damage and genotoxicity, and DSBs were quantified by γH2AX-foci-staining using confocal microscopy. Flow cytometry analysis was done to determine the cell cycle, mitochondrial membrane depolarization, and Reactive oxygen species generation. Pharmacophore analysis and protein-ligand interaction studies were done using Schrodinger software. Synergy was computed by compusyn-statistical analysis. Standard errors/deviation/significance were computed with GraphPad prism. RESULTS Using piperine, we propose a new strategy for overcoming acquired radioresistance through estrogen receptor-mediated modulation of the NHEJ pathway. This is the first comprehensive study elucidating the mechanism of radio sensitizing potential of piperine. Piperine enhanced the radiation-induced cell death and enhanced the expression and activation of Estrogen receptor β, while Estrogen receptor α expression and activation were reduced. In addition, piperine shares common pharmacophore features with most of the known estrogen agonists and antagonists. It altered the estrogen receptor α/β ratio and the expression of estrogen-responsive proteins of DDR and NHEJ pathway. Enhanced expression of DDR proteins, ATM, p53, and P-p53 with low DNA-PK repair complex (comprising of DNA-PKcs/Ku70/Ku80), resulted in the accumulation of radiation-induced DNA double-stranded breaks (as evidenced by MNi and γH2AX-foci) culminating in cell cycle arrest and mitochondrial-pathway of apoptosis. CONCLUSION In conclusion, our study for the first time reported that piperine sensitizes breast cancer cells to radiation by accumulating DNA breaks, through altering the expression of DNA-PK Complex, and DDR proteins, via selective estrogen receptor modulation, offering a novel strategy for combating radioresistance.
Collapse
Affiliation(s)
- Koniyan Shaheer
- Division of Cancer Research and Therapeutics (CaRT), Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - Br Swathi Prabhu
- Division of Cancer Research and Therapeutics (CaRT), Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - H Shabeer Ali
- Department of Biotechnology and Microbiology, Kannur University, Kannur, Kerala, India
| | - Divya Lakshmanan-M
- Division of Cancer Research and Therapeutics (CaRT), Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India.
| |
Collapse
|
14
|
Ishitani T. Cadherin-linked morphogen gradient actualizes robust tissue patterning. Curr Opin Cell Biol 2023; 85:102275. [PMID: 37944424 DOI: 10.1016/j.ceb.2023.102275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/30/2023] [Accepted: 10/14/2023] [Indexed: 11/12/2023]
Abstract
Morphogen gradients govern tissue patterning. These gradients provide positional information, instructing cells to adopt distinct fates. Over the past few decades, extensive studies have revealed the detailed mechanisms by which morphogens generate tissue patterns. However, the communication between morphogen-receiving cells is still poorly understood. Here, I describe how cadherin-mediated cell competition ensures robust morphogen-gradient formation. In normal zebrafish embryos, unfit cells with abnormal Wnt signaling activity spontaneously appear and produce a noisy morphogen gradient. These unfit cells communicate with neighboring cells through cadherins and are subsequently killed by cell competition. This process of killing unfit cells corrects noisy gradients to support reproducible patterning. I also discuss the significance of cell-competition-mediated morphogen-gradient correction from the perspectives of evolution and disease biology.
Collapse
Affiliation(s)
- Tohru Ishitani
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan; Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
15
|
Zhang L, Shi L, Han J, Li Z. Protection of β-pancreatic cells from dysfunctionality of insulin using vitexin by apoptosis of INS-1 cells. Arch Physiol Biochem 2023; 129:1160-1167. [PMID: 33835897 DOI: 10.1080/13813455.2021.1910714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
AIMS This study was performed to explore the possible beneficial effects of vitexin on high glucose (HG)-induced cytotoxicity in pancreatic β-cells. METHODS INS-1 pancreatic β-cell line has used this study. HG-induced (33 Mm) exposed INS-1 cell death; the apoptosis INS-1 cells treated vitexin 10, 20, 40, and 80 µg/mL for 24 hours. The anti-apoptosis properties were evaluated by MTT assay, glucose-stimulated insulin secretion assay, biochemical assay, annexin-V-FITC staining and western blot analysis. RESULTS These findings demonstrate that vitexin treatment improved the HG-exposure, reduced the INS-1 cell viability and significantly enhanced glucose-stimulated insulin secretion in a dose-dependent manner. The antioxidant studies revealed that vitexin treatment significantly decreased lipid peroxidation and reactive oxygen species and increased antioxidant level of INS-1 cell line in 24 hrs. The findings of the study suggested that in the vitexin treatment group, pancreatic apoptosis and Bax protein expression reduced significantly. At the same time, Bcl-2 protein expression increased, and NF-κB protein in HG-induced INS-cells was inhibited. CONCLUSION Therefore, our results suggest that vitexin can be successfully used to regulate the expression of Bcl-2 family proteins, reduce lipid peroxidation and to improve the secretion of antioxidants in pancreatic β-cell lines.
Collapse
Affiliation(s)
- Li Zhang
- Department of endocrinology, The Fourth People's Hospital of Jinan city, Jinan, Shandong Province, China
| | - Lianfeng Shi
- Department of First General Medicine, Binzhou People's Hospital, Binzhou, Shandong Province, China
| | - Juanjuan Han
- Department of First General Medicine, Binzhou People's Hospital, Binzhou, Shandong Province, China
| | - Zhenzuo Li
- Department of endocrinology, The Fourth People's Hospital of Jinan city, Jinan, Shandong Province, China
| |
Collapse
|
16
|
He Y, Zheng J, Ye B, Dai Y, Nie K. Chemotherapy-induced gastrointestinal toxicity: Pathogenesis and current management. Biochem Pharmacol 2023; 216:115787. [PMID: 37666434 DOI: 10.1016/j.bcp.2023.115787] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Chemotherapy is the most common treatment for malignant tumors. However, chemotherapy-induced gastrointestinal toxicity (CIGT) has been a major concern for cancer patients, which reduces their quality of life and leads to treatment intolerance and even cessation. Nevertheless, prevention and treatment for CIGT are challenging, due to the prevalence and complexity of the condition. Chemotherapeutic drugs directly damage gastrointestinal mucosa to induce CIGT, including nausea, vomiting, anorexia, gastrointestinal mucositis, and diarrhea, etc. The pathogenesis of CIGT involves multiple factors, such as gut microbiota disorders, inflammatory responses and abnormal neurotransmitter levels, that synergistically contribute to its occurrence and development. In particular, the dysbiosis of gut microbiota is usually linked to abnormal immune responses that increases inflammatory cytokines' expression, which is a common characteristic of many types of CIGT. Chemotherapy-induced intestinal neurotoxicity is also a vital concern in CIGT. Currently, modern medicine is the dominant treatment of CIGT, however, traditional Chinese medicine (TCM) has attracted interest as a complementary and alternative therapy that can greatly alleviate CIGT. Accordingly, this review aimed to comprehensively summarize the pathogenesis and current management of CIGT using PubMed and Google Scholar databases, and proposed that future research for CIGT should focus on the gut microbiota, intestinal neurotoxicity, and promising TCM therapies, which may help to develop more effective interventions and optimize managements of CIGT.
Collapse
Affiliation(s)
- Yunjing He
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jingrui Zheng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Binbin Ye
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yongzhao Dai
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ke Nie
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
17
|
Simón L, Arazo-Rusindo M, Quest AFG, Mariotti-Celis MS. Phlorotannins: Novel Orally Administrated Bioactive Compounds That Induce Mitochondrial Dysfunction and Oxidative Stress in Cancer. Antioxidants (Basel) 2023; 12:1734. [PMID: 37760037 PMCID: PMC10525198 DOI: 10.3390/antiox12091734] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Mitochondrial dysfunction is an interesting therapeutic target to help reduce cancer deaths, and the use of bioactive compounds has emerged as a novel and safe approach to solve this problem. Here, we discuss the information available related to phlorotannins, a type of polyphenol present in brown seaweeds that reportedly functions as antioxidants/pro-oxidants and anti-inflammatory and anti-tumorigenic agents. Specifically, available evidence indicates that dieckol and phloroglucinol promote mitochondrial membrane depolarization and mitochondria-dependent apoptosis. Phlorotannins also reduce pro-tumorigenic, -inflammatory, and -angiogenic signaling mechanisms involving RAS/MAPK/ERK, PI3K/Akt/mTOR, NF-κB, and VEGF. In doing so, they inhibit pathways that favor cancer development and progression. Unfortunately, these compounds are rather labile and, therefore, this review also summarizes approaches permitting the encapsulation of bioactive compounds, like phlorotannins, and their subsequent oral administration as novel and non-invasive therapeutic alternatives for cancer treatment.
Collapse
Affiliation(s)
- Layla Simón
- Nutrition and Dietetic School, Facultad de Medicina, Universidad Finis Terrae, Santiago 7501015, Chile
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile;
| | - Migdalia Arazo-Rusindo
- Department of Chemical and Bioprocess Engineering, Faculty of Engineering, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile;
| | - Andrew F. G. Quest
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile
| | | |
Collapse
|
18
|
Zhou X, An B, Lin Y, Ni Y, Zhao X, Liang X. Molecular mechanisms of ROS-modulated cancer chemoresistance and therapeutic strategies. Biomed Pharmacother 2023; 165:115036. [PMID: 37354814 DOI: 10.1016/j.biopha.2023.115036] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023] Open
Abstract
Drug resistance is the main obstacle to achieving a cure in many cancer patients. Reactive oxygen species (ROS) are master regulators of cancer development that act through complex mechanisms. Remarkably, ROS levels and antioxidant content are typically higher in drug-resistant cancer cells than in non-resistant and normal cells, and have been shown to play a central role in modulating drug resistance. Therefore, determining the underlying functions of ROS in the modulation of drug resistance will contribute to develop therapies that sensitize cancer resistant cells by leveraging ROS modulation. In this review, we summarize the notable literature on the sources and regulation of ROS production and highlight the complex roles of ROS in cancer chemoresistance, encompassing transcription factor-mediated chemoresistance, maintenance of cancer stem cells, and their impact on the tumor microenvironment. We also discuss the potential of ROS-targeted therapies in overcoming tumor therapeutic resistance.
Collapse
Affiliation(s)
- Xiaoting Zhou
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Biao An
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yi Lin
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yanghong Ni
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xiao Liang
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
19
|
Badr AM, El-Orabi NF, Mahran YF, Badr AM, Bayoumy NM, Hagar H, Elmongy EI, Atawia RT. In vivo and In silico evidence of the protective properties of carvacrol against experimentally-induced gastric ulcer: Implication of antioxidant, anti-inflammatory, and antiapoptotic mechanisms. Chem Biol Interact 2023; 382:110649. [PMID: 37499997 DOI: 10.1016/j.cbi.2023.110649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Gastric ulcer is a serious disease that affects millions of individuals worldwide. Alcohol consumption is a major contributor to the disease pathogenesis and ethanol-induced ulcer in rats closely recapitulates the clinical pathology of ulcer. In this study, rats were pretreated with carvacrol (CAR,50 and 100 mg/kg, orally) 1 h before absolute ethanol administration to induce gastric ulcer. CAR prevented ethanol-induced increases in gastric volume and acidity while restored mucin content. The gastro-protective activity of CAR, particularly the higher dose (100 mg/kg), was further supported by histopathological examination, as manifested by reduced gastric lesions. Interestingly, oxidative stress is linked to early stages of ulcer development and progression. In this study, ethanol administration upregulated the levels of ROS-producing enzymes, NADPH oxidase homologs 1 and 4 (Nox1 and Nox4) and lipid peroxides while depleting the antioxidant defense mechanisms, including GSH, Glutathione Peroxidase (GPX) and catalase. Interestingly, these alterations were significantly ameliorated by CAR pretreatment. Additionally, CAR possesses anti-inflammatory and anti-apoptotic activities. Pretreatment with CAR blunted ethanol-induced increases in inflammatory cytokines (NF-κB and TNF-α) and rectified the apoptosis regulator (Bax/Bcl2 ratio) in gastric tissue. Moreover, the docking simulation of CAR illustrated good fitting and interactions with GPX, Nox1 and TNF-α through the formation of hydrogen and hydrophobic (pi-H) bonds with conservative amino acids, thus, further supporting the anti-inflammatory and antioxidant effects underlying the gastroprotective effects of CAR. In conclusion, this study elucidates, using in silico and in vivo models, that the gastroprotective activity of CAR is attributed, at least in part, to its mucin-secretagogue, antioxidative, anti-inflammatory, and anti-apoptotic mechanisms.
Collapse
Affiliation(s)
- Amira M Badr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Naglaa F El-Orabi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Yasmen F Mahran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abasia, Cairo, Egypt
| | - Amul M Badr
- Department of Medical Biochemistry and Molecular Biology, Kasr Al-Aini Faculty of Medicine, Cairo University, Egypt
| | | | - Hanan Hagar
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Elshaymaa I Elmongy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo, Egypt
| | - Reem T Atawia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abasia, Cairo, Egypt; Department of Pharmaceutical Sciences, College of Pharmacy, Southwestern Oklahoma State University, Weatherford, OK, United States.
| |
Collapse
|
20
|
Wang K, Che W, Duan M, Wang C, Li X, He L. Effects of Broflanilide on Oxidative Stress and Expression of Apoptotic Genes in Zebrafish (Danio rerio) Gill. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2023; 110:91. [PMID: 37156957 DOI: 10.1007/s00128-023-03733-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 04/20/2023] [Indexed: 05/10/2023]
Abstract
Broflanilide exerted negative impacts on the gill of zebrafish. Thus, in this study, zebrafish gill was used to assess the apoptosis toxicity of broflanilide by determining the levels of reactive oxygen species (ROS), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and malondialdehyde (MDA) and apoptosis-related genes. The results found that the minimum threshold for the content and time of broflanilide affecting enzyme content and gene expression was 0.26 mg/L after 24 h exposure. After 96 h exposure, broflanilide could cause apoptosis and exerted significantly increased contents of ROS and MDA, while inhibiting the activities of SOD, CAT, and GPx at 0.26 and 0.57 mg/L. Broflanilide also had adverse effects on apoptosis-related genes, such as tumor protein p53 (p53), associated × (Bax), B-cell lymphama-2 (Bcl-2), caspase-3, caspase-9, and apoptotic protease activating factor-1(apaf-1), at 0.26 mg/L and 0.57 mg/L after 96 h exposure, respectively. These results provide new insight into the potential toxicity mechanisms of broflanilide in zebrafish gills.
Collapse
Affiliation(s)
- Kai Wang
- Plant Protection College, Shenyang Agricultural University, Shenyang, China.
| | - Wunan Che
- Plant Protection College, Shenyang Agricultural University, Shenyang, China
| | - Manman Duan
- College of Science, China Agricultural University, Beijing, China
| | - Chengju Wang
- College of Science, China Agricultural University, Beijing, China
| | - Xiuwei Li
- Plant Protection College, Shenyang Agricultural University, Shenyang, China
| | - Lu He
- Plant Protection College, Shenyang Agricultural University, Shenyang, China.
| |
Collapse
|
21
|
Hassab LY, Abbas SS, Mohammed RA, Abdallah DM. Dimethyl fumarate abrogates striatal endoplasmic reticulum stress in experimentally induced late-stage Huntington’s disease: Focus on the IRE1α/JNK and PERK/CHOP trajectories. Front Pharmacol 2023; 14:1133863. [PMID: 37056990 PMCID: PMC10088517 DOI: 10.3389/fphar.2023.1133863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Introduction: Dimethyl fumarate (DMF) is FDA-approved for use in patients with relapsing multiple sclerosis, and it processes neuroprotection in several experimental settings; however, its impact on combating Huntington’s disease (HD) remains elusive. This study aimed to explore the role of DMF post-treatment on HD mediated endoplasmic reticulum (ER) stress response in a selective striatal degeneration HD model.Methods: Rats, exposed to 3-nitropropionic acid, were either left untreated or post-treated with DMF for 14 days.Results and Discussion: DMF reduced locomotion deficits in both the open field and beam walk paradigms, boosted the striatal dopamine (DA) content, improved its architecture at the microscopic level, and hindered astrogliosis. Mechanistically, DMF limited the activation of two of the ER stress arms in the striatum by reducing p-IRE1α, p-JNK, and p-PERK protein expressions besides the CHOP/GADD153 content. Downstream from both ER stress arms’ suppression, DMF inhibited the intrinsic apoptotic pathway, as shown by the decrease in Bax and active caspase-3 while raising Bcl-2. DMF also decreased oxidative stress markers indicated by a decline in both reactive oxygen species and malondialdehyde while boosting glutathione. Meanwhile, it enhanced p-AKT to activate /phosphorylate mTOR and stimulate the CREB/BDNF/TrkB trajectory, which, in a positive feedforward loop, activates AKT again. DMF also downregulated the expression of miRNA-634, which negatively regulates AKT, to foster survival kinase activation.Conclusion: This study features a focal novel point on the DMF therapeutic ability to reduce HD motor manifestations via its ability to enhance DA and suppress the IRE1α/JNK and PERK/CHOP/GADD153 hubs to inhibit the mitochondrial apoptotic pathway through activating the AKT/mTOR and BDNF/TrkB/AKT/CREB signaling pathways and abating miRNA-634 and oxidative stress.
Collapse
Affiliation(s)
- Lina Y. Hassab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Samah S. Abbas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Reham A. Mohammed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Dalaal M. Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- *Correspondence: Dalaal M. Abdallah,
| |
Collapse
|
22
|
Rai N, Gupta P, Verma A, Tiwari RK, Madhukar P, Kamble SC, Kumar A, Kumar R, Singh SK, Gautam V. Ethyl Acetate Extract of Colletotrichum gloeosporioides Promotes Cytotoxicity and Apoptosis in Human Breast Cancer Cells. ACS OMEGA 2023; 8:3768-3784. [PMID: 36743019 PMCID: PMC9893742 DOI: 10.1021/acsomega.2c05746] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/11/2023] [Indexed: 06/18/2023]
Abstract
Fungal endophytes are known to be a paragon for producing bioactive compounds with a variety of pharmacological importance. The current study aims to elucidate the molecular alterations induced by the bioactive compounds produced by the fungal endophyte Colletotrichum gloeosporioides in the tumor microenvironment of human breast cancer cells. GC/MS analysis of the ethyl acetate (EA) extract of C. gloeosporioides revealed the presence of bioactive compounds with anticancer activity. The EA extract of C. gloeosporioides exerted potential plasmid DNA protective activity against hydroxyl radicals of Fenton's reagent. The cytotoxic activity further revealed that MDA-MB-231 cells exhibit more sensitivity toward the EA extract of C. gloeosporioides as compared to MCF-7 cells, whereas non-toxic to non-cancerous HEK293T cells. Furthermore, the anticancer activity demonstrated by the EA extract of C. gloeosporioides was studied by assessing nuclear morphometric analysis and induction of apoptosis in MDA-MB-231 and MCF-7 cells. The EA extract of C. gloeosporioides causes the alteration in cellular and nuclear morphologies, chromatin condensation, long-term colony inhibition, and inhibition of cell migration and proliferation ability of MDA-MB-231 and MCF-7 cells. The study also revealed that the EA extract of C. gloeosporioides treated cells undergoes apoptosis by increased production of reactive oxygen species and significant deficit in mitochondrial membrane potential. Our study also showed that the EA extract of C. gloeosporioides causes upregulation of pro-apoptotic (BAX, PARP, CASPASE-8, and FADD), cell cycle arrest (P21), and tumor suppressor (P53) related genes. Additionally, the downregulation of antiapoptotic genes (BCL-2 and SURVIVIN) and increased Caspase-3 activity suggest the induction of apoptosis in the EA extract of C. gloeosporioides treated MDA-MB-231 and MCF-7 cells. Overall, our findings suggest that the bioactive compounds present in the EA extract of C. gloeosporioides promotes apoptosis by altering the genes related to the extrinsic as well as the intrinsic pathway. Further in vivo study in breast cancer models is required to validate the in vitro observations.
Collapse
Affiliation(s)
- Nilesh Rai
- Centre
of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Priyamvada Gupta
- Centre
of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Ashish Verma
- Centre
of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Rajan Kumar Tiwari
- Department
of Zoology, Institute of Science, Banaras
Hindu University, Varanasi, 221005, India
| | - Prasoon Madhukar
- Infectious
Disease Research Laboratory, Department of Medicine, Institute of
Medical Sciences, Banaras Hindu University, Varanasi221005, India
| | - Swapnil C. Kamble
- Department
of Technology, Savitribai Phule Pune University, Ganeshkhind, Pune, 411007, India
| | - Ajay Kumar
- Department
of Zoology, Institute of Science, Banaras
Hindu University, Varanasi, 221005, India
| | - Rajiv Kumar
- Centre
of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Santosh Kumar Singh
- Centre
of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Vibhav Gautam
- Centre
of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
23
|
Li Y, Zhang X, Wang Z, Li B, Zhu H. Modulation of redox homeostasis: A strategy to overcome cancer drug resistance. Front Pharmacol 2023; 14:1156538. [PMID: 37033606 PMCID: PMC10073466 DOI: 10.3389/fphar.2023.1156538] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Cancer treatment is hampered by resistance to conventional therapeutic strategies, including chemotherapy, immunotherapy, and targeted therapy. Redox homeostasis manipulation is one of the most effective innovative treatment techniques for overcoming drug resistance. Reactive oxygen species (ROS), previously considered intracellular byproducts of aerobic metabolism, are now known to regulate multiple signaling pathways as second messengers. Cancer cells cope with elevated amounts of ROS during therapy by upregulating the antioxidant system, enabling tumor therapeutic resistance via a variety of mechanisms. In this review, we aim to shed light on redox modification and signaling pathways that may contribute to therapeutic resistance. We summarized the molecular mechanisms by which redox signaling-regulated drug resistance, including altered drug efflux, action targets and metabolism, enhanced DNA damage repair, maintained stemness, and reshaped tumor microenvironment. A comprehensive understanding of these interrelationships should improve treatment efficacy from a fundamental and clinical research point of view.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Biotherapy and Cancer Center, West China School of Basic Medical Sciences and Forensic Medicine, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xiaoyue Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China School of Basic Medical Sciences and Forensic Medicine, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Zhihan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China School of Basic Medical Sciences and Forensic Medicine, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China School of Basic Medical Sciences and Forensic Medicine, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Huili Zhu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
- *Correspondence: Huili Zhu,
| |
Collapse
|
24
|
Bai J, Wu L, Wang X, Wang Y, Shang Z, Jiang E, Shao Z. Roles of Mitochondria in Oral Squamous Cell Carcinoma Therapy: Friend or Foe? Cancers (Basel) 2022; 14:cancers14235723. [PMID: 36497206 PMCID: PMC9738284 DOI: 10.3390/cancers14235723] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) therapy is unsatisfactory, and the prevalence of the disease is increasing. The role of mitochondria in OSCC therapy has recently attracted increasing attention, however, many mechanisms remain unclear. Therefore, we elaborate upon relative studies in this review to achieve a better therapeutic effect of OSCC treatment in the future. Interestingly, we found that mitochondria not only contribute to OSCC therapy but also promote resistance, and targeting the mitochondria of OSCC via nanoparticles is a promising way to treat OSCC.
Collapse
Affiliation(s)
- Junqiang Bai
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
| | - Luping Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
| | - Xinmiao Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
| | - Yifan Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
| | - Zhengjun Shang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
| | - Erhui Jiang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
- Correspondence: (E.J.); (Z.S.); Tel.: +86-27-87686215 (E.J. & Z.S.)
| | - Zhe Shao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430089, China
- Correspondence: (E.J.); (Z.S.); Tel.: +86-27-87686215 (E.J. & Z.S.)
| |
Collapse
|
25
|
Huang B, Lang X, Li X. The role of TIGAR in nervous system diseases. Front Aging Neurosci 2022; 14:1023161. [DOI: 10.3389/fnagi.2022.1023161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022] Open
Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) mainly regulates pentose phosphate pathway by inhibiting glycolysis, so as to synthesize ribose required by DNA, promote DNA damage repair and cell proliferation, maintain cell homeostasis and avoid body injury. Its physiological functions include anti-oxidative stress, reducing inflammation, maintaining mitochondrial function, inhibiting apoptosis, reducing autophagy etc. This paper reviews the research of TIGAR in neurological diseases, including stroke, Parkinson’s disease (PD), Alzheimer’s disease (AD), seizures and brain tumors, aiming to provide reference for the development of new therapeutic targets.
Collapse
|
26
|
George BP, Rajendran NK, Houreld NN, Abrahamse H. Rubus Capped Zinc Oxide Nanoparticles Induce Apoptosis in MCF-7 Breast Cancer Cells. Molecules 2022; 27:molecules27206862. [PMID: 36296460 PMCID: PMC9611499 DOI: 10.3390/molecules27206862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/28/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Rubus fairholmianus (RF) has widely been used to treat various ailments, including pain, diabetes, and cancer. Zinc oxide nanoparticles (ZnO NPs) have drawn attention in modern healthcare applications. Hence, we designed this study to synthesize zinc oxide (ZnO) nanoparticles using R. fairholmianus root extract to investigate its synergistic cytotoxic effect on MCF-7 cells and explore the possible cell death mechanism. ZnO NPs were synthesized via green synthesis using R. fairholmianus root extract, and the effect on MCF-7 cells was determined by looking at cellular morphology, proliferation, cytotoxicity, apoptosis, and reactive oxygen species (ROS). The results showed that cellular proliferation was reduced following treatment with R. fairholmianus capped zinc oxide nanoparticles (RFZnO NPs), while cytotoxicity and ROS were increased. There was also an increase in apoptosis as indicated by the significant increase in cytoplasmic cytochrome c and caspase 3/7 (markers of apoptosis), as well as increased levels of pro-apoptotic proteins (p53, Bax) and decreased levels of anti-apoptotic protein (Bcl-2). In conclusion, these results showed that RFZnO NPs induce apoptosis in breast cancer cells via a mitochondria-mediated caspase-dependent apoptotic pathway and suggest the use of acetone root extract of R. fairholmianus for the treatment of cancer-related ailments.
Collapse
|
27
|
Zhang Y, Liu Z, Wang X, Jian H, Xiao H, Wen T. SHMT2 promotes cell viability and inhibits ROS-dependent, mitochondrial-mediated apoptosis via the intrinsic signaling pathway in bladder cancer cells. Cancer Gene Ther 2022; 29:1514-1527. [PMID: 35422087 DOI: 10.1038/s41417-022-00470-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/16/2022] [Accepted: 03/30/2022] [Indexed: 11/08/2022]
Abstract
Mitochondrial serine hydroxymethyltransferase (SHMT2) catalyzes the conversion of serine to glycine and concomitantly produces one-carbon units to support cell growth and is upregulated in various cancer cells. SHMT2 knockdown triggers cell apoptosis; however, the detailed mechanism of apoptosis induced by SHMT2 inactivation remains unknown. Here, we demonstrate that SHMT2 supports the proliferation of bladder cancer (BC) cells by maintaining redox homeostasis. SHMT2 knockout decreased the pools of purine and one-carbon units and delayed cell cycle progression in a manner that was rescued by formate, demonstrating that SHMT2-mediated one-carbon units are essential for BC cell proliferation. SHMT2 deficiency promoted the accumulation of intracellular reactive oxygen species (ROS) by decreasing the NADH/NAD+, NADPH/NADP+, and GSH/GSSG ratios, leading to a loss in mitochondrial membrane potential, release of cytochrome c, translocation of Bcl-2 family protein and activation of caspase-3. Notably, blocking ROS production with the one-carbon donor formate and the ROS scavenger N-acetyl-cysteine (NAC) effectively rescued SHMT2 deficiency-induced cell apoptosis via the intrinsic signaling pathway. Treatment with the SHMT inhibitor SHIN1 resulted in a significant inhibitory effect on cell proliferation and induced cell apoptosis. Formate and NAC rescued SHIN1-induced cell apoptosis. Our findings reveal an important mechanism by which the loss of SHMT2 triggers ROS-dependent, mitochondrial-mediated apoptosis, which gives insight into the link between serine metabolism and cell apoptosis and provides a promising target for BC treatment and drug discovery.
Collapse
Affiliation(s)
- Yun Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, 100190, Beijing, China
| | - Zhe Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Xueliang Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Hui Jian
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Haihan Xiao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Tingyi Wen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China.
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, 100190, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
28
|
WalyEldeen AA, El-Shorbagy HM, Hassaneen HM, Abdelhamid IA, Sabet S, Ibrahim SA. [1,2,4] Triazolo [3,4-a]isoquinoline chalcone derivative exhibits anticancer activity via induction of oxidative stress, DNA damage, and apoptosis in Ehrlich solid carcinoma-bearing mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1225-1238. [PMID: 35881165 PMCID: PMC9467967 DOI: 10.1007/s00210-022-02269-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 07/03/2022] [Indexed: 12/30/2022]
Abstract
Despite the advances made in cancer therapeutics, their adverse effects remain a major concern, putting safer therapeutic options in high demand. Since chalcones, a group of flavonoids and isoflavonoids, act as promising anticancer agents, we aimed to evaluate the in vivo anticancer activity of a synthetic isoquinoline chalcone (CHE) in a mice model with Ehrlich solid carcinoma. Our in vivo pilot experiments revealed that the maximum tolerated body weight-adjusted CHE dose was 428 mg/kg. Female BALB/c mice were inoculated with Ehrlich ascites carcinoma cells and randomly assigned to three different CHE doses administered intraperitoneally (IP; 107, 214, and 321 mg/kg) twice a week for two consecutive weeks. A group injected with doxorubicin (DOX; 4 mg/kg IP) was used as a positive control. We found that in CHE-treated groups: (1) tumor weight was significantly decreased; (2) the total antioxidant concentration was substantially depleted in tumor tissues, resulting in elevated oxidative stress and DNA damage evidenced through DNA fragmentation and comet assays; (3) pro-apoptotic genes p53 and Bax, assessed via qPCR, were significantly upregulated. Interestingly, CHE treatment reduced immunohistochemical staining of the proliferative marker ki67, whereas BAX was increased. Notably, histopathological examination indicated that unlike DOX, CHE treatment had minimal toxicity on the liver and kidney. In conclusion, CHE exerts antitumor activity via induction of oxidative stress and DNA damage that lead to apoptosis, making CHE a promising candidate for solid tumor therapy.
Collapse
Affiliation(s)
| | - Haidan M El-Shorbagy
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
- Faculty of Biotechnology, October University for Modern Science and Art (MSA), 6th October, Cairo, Egypt
| | - Hamdi M Hassaneen
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Ismail A Abdelhamid
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Salwa Sabet
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | | |
Collapse
|
29
|
Reactive oxygen species, the trident of Neptune in the hands of hecate; role in different diseases, signaling pathways, and detection methods. Arch Biochem Biophys 2022; 728:109357. [DOI: 10.1016/j.abb.2022.109357] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/29/2022] [Accepted: 07/16/2022] [Indexed: 12/22/2022]
|
30
|
Gong J, Li Q, Yu H, Liu S, Kong L. Effects of low salinity on hemolymph osmolality and transcriptome of the Iwagaki oyster, Crassostrea nippona. FISH & SHELLFISH IMMUNOLOGY 2022; 126:211-216. [PMID: 35636697 DOI: 10.1016/j.fsi.2022.05.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/06/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
Crassostrea nippona is a kind of oysters with great development value as it can be edible in summer for its late reproductive period. Salinity is one of the important limiting abiotic factors to the survival and distribution of this stenohaline species. To better understand the physiological and immunological response of C. nippona to varying environmental salinities, the effects of low salinity on the hemolymph osmolality and gill transcriptome were investigated in this study. The osmolality of hemolymph in vivo and surrounding water were assessed regularly over one week at five test salinities ranging from 5 psμ to 30 psμ. They reached osmotic equilibrium within hours above 15 psμ but remained hyperosmotic at 10 and 5 psμ for the whole sampling period. Through comparative transcriptome analysis, there were less differentially expressed genes (DEGs) in pairwise comparison of S1 (10 psμ) vs S3 (30 psμ) than in S2 (20 psμ) vs S3. KEGG enrichment analysis identified ubiquitin-mediated proteolysis and mitochondrial apoptosis pathway specifically enriched at 10 psμ. This study gained comprehensive insights on the low salinity response of C. nippona at the molecular level, which provide a theoretical basis for understanding the immune mechanism under low salinity stress.
Collapse
Affiliation(s)
- Jianwen Gong
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 66003, China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 66003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| | - Hong Yu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 66003, China
| | - Shikai Liu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 66003, China
| | - Lingfeng Kong
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 66003, China
| |
Collapse
|
31
|
Phyllostachys nigra Variety Henosis, a Domestic Bamboo Species, Protects PC12 Cells from Oxidative Stress-mediated Cell Injury through Nrf2 Activation. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-021-0395-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Jaiswara PK, Kumar A. Nimbolide retards T cell lymphoma progression by altering apoptosis, glucose metabolism, pH regulation, and ROS homeostasis. ENVIRONMENTAL TOXICOLOGY 2022; 37:1445-1457. [PMID: 35199915 DOI: 10.1002/tox.23497] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/05/2022] [Accepted: 02/13/2022] [Indexed: 06/14/2023]
Abstract
Nimbolide is reported as one of the potential anticancer candidates of the neem tree (Azadirachta indica A. Juss). The cytotoxic action of nimbolide has been well reported against a wide number of malignancies, including breast, prostate, lung, liver, and cervix cancers. Interestingly, only a few in vivo studies conducted on B cell lymphoma, glioblastoma, pancreatic cancer, and buccal pouch carcinoma have shown the in vivo antitumor efficacy of nimbolide. Therefore, it is highly needed to examine the in vivo antineoplastic activity of nimbolide on a wide variety of cancers to establish nimbolide as a promising anticancer drug. In the present study, we investigated the tumor retarding action of nimbolide in a murine model of T cell lymphoma. We noticed significantly augmented apoptosis in nimbolide- administered tumor-bearing mice, possibly due to down-regulated expression of Bcl2 and up-regulated expression of p53, cleaved caspase-3, Cyt c, and ROS. The nimbolide treatment-induced ROS production by suppressing the expression of antioxidant regulatory enzymes, namely superoxide dismutase and catalase. In addition, nimbolide administration impaired glycolysis and pH homeostasis with concomitant inhibition of crucial glycolysis and pH regulatory molecules such as GLUT3, LDHA, MCT1, and V-ATPase, CAIX and NHE1, respectively. Taken together, the present investigation provides novel insights into molecular mechanisms of nimbolide inhibited T cell lymphoma progression and directs the utility of nimbolide as a potential anticancer therapeutic drug for the treatment of T cell lymphoma.
Collapse
Affiliation(s)
- Pradip Kumar Jaiswara
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ajay Kumar
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
33
|
Tanwar S, Thakur V, Parsad D. Dopamine toxicity contributes to melanocyte loss via melanocytorrhagy: an in vitro study. Int J Dermatol 2022; 61:1253-1261. [PMID: 35325470 DOI: 10.1111/ijd.16166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/27/2021] [Accepted: 03/03/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Catecholamines (epinephrine, norepinephrine, and dopamine) have been proposed as a possible cause of melanocyte loss. Dopamine has been observed to cause apoptosis in melanocytes via reactive oxygen species development, but the effect on melanocyte adhesion and proliferation still remains to be elucidated. Thus, we explored the dose- and time-dependent toxicity of catecholamines and the effect of dopamine on the proliferation and adhesion potential of melanocytes. MATERIALS AND METHODS Primary culture of melanocytes was investigated in vitro for toxic effects of epinephrine, norepinephrine, and dopamine on metabolic activity via MTT [3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide) assay. Cells were observed microscopically for any phenotypic changes. Further cell proliferation, cell adhesion, and cell death pathway were explored under dopamine toxicity in dose- and time-dependent manner with RNase/PI method of cell cycle analysis, cell adhesion assay, and Annexin V-FITC/PI assay, respectively. Altered gene expressions were confirmed with a real-time polymerase chain reaction. RESULTS Metabolic activity of cells varied with time and different doses of epinephrine, norepinephrine, and dopamine. Dopamine was observed to be more toxic than epinephrine and norepinephrine. Melanocytes were observed to follow different cell death pathways at comparatively lower and higher concentrations of dopamine. Persistent exposure to dopamine resulted in decreased cell proliferation and adhesion potential with apoptotic changes. Gene expression changes also confirmed the weak adhesion and survival potential of cells under the toxic effects of dopamine. CONCLUSION Dopamine can alter melanocytes' adhesion and survival potential, leading to apoptotic cell death or melanocytorrhagic loss.
Collapse
Affiliation(s)
- Sushma Tanwar
- Department of Dermatology, Venereology and Leprology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Vishal Thakur
- Department of Dermatology, Venereology and Leprology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Davinder Parsad
- Department of Dermatology, Venereology and Leprology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
34
|
Changizi V, Azadbakht O, Ghanavati R, Behrouj H, Motevaseli E, Khanzadeh P. Effect of Lactobacillus species on apoptosis-related genes BCL2, BAX, and caspase 3 in the testes of gamma-irradiated rats. Rev Assoc Med Bras (1992) 2021; 67:1581-1585. [PMID: 34909882 DOI: 10.1590/1806-9282.20210634] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/30/2023] Open
Abstract
OBJECTIVE Ionizing radiation has various applications, including uses in medicine, industry, agriculture, and research. However, ionizing radiation is accompanied by side effects in normal radiosensitive tissues. Probiotics as natural radioprotective agents can protect normal tissues from ionizing radiation. In this regard, this study aimed to investigate the effect of Lactobacillus species on apoptosis-related genes BCL2, BAX, and caspase 3 (CASP3) in the testes of gamma-irradiated rats. METHODS A total of 30 male Wistar rats were involved in this study. The animals received the whole- body radiation with the dose rate of 2 Gy gamma-ray and were orally gavaged with 0.2 mL of 1×1010 Lactobacillus species in phosphate-buffered saline (PBS) for 4 weeks. Then, the relative gene expression levels of BCL2, BAX, and CASP3 in the testis were assessed by using the quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS Compared with the control group, radiation significantly downregulated the BCL2 and upregulated the BAX and CASP3 genes (p<0.0001). However, Lactobacillus species significantly reversed these effects. CONCLUSION All in all, according to our results, employing Lactobacilli probiotics as a natural radioprotector may protect radiosensitive tissue from damage.
Collapse
Affiliation(s)
- Vahid Changizi
- Tehran University of Medical Sciences, Allied Medical Sciences School, Technology of Radiology and Radiotherapy Department - Tehran, Iran
| | - Omid Azadbakht
- Behbahan Faculty of Medical Sciences, Radiology Technology Department - Behbahan, Iran
| | - Roya Ghanavati
- Behbahan Faculty of Medical Sciences, Medical Laboratory Sciences Department - Behbahan, Iran
| | - Hamid Behrouj
- Behbahan Faculty of Medical Sciences, Medical Laboratory Sciences Department - Behbahan, Iran
| | - Elahe Motevaseli
- Tehran University of Medical Sciences, Advanced Technologies in Medicine School, Molecular Medicine Department - Tehran, Iran
| | - Pegah Khanzadeh
- Tehran University of Medical Sciences, Allied Medical Sciences School, Technology of Radiology and Radiotherapy Department - Tehran, Iran
| |
Collapse
|
35
|
Brahma MK, Gilglioni EH, Zhou L, Trépo E, Chen P, Gurzov EN. Oxidative stress in obesity-associated hepatocellular carcinoma: sources, signaling and therapeutic challenges. Oncogene 2021; 40:5155-5167. [PMID: 34290399 PMCID: PMC9277657 DOI: 10.1038/s41388-021-01950-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/01/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023]
Abstract
Obesity affects more than 650 million individuals worldwide and is a well-established risk factor for the development of hepatocellular carcinoma (HCC). Oxidative stress can be considered as a bona fide tumor promoter, contributing to the initiation and progression of liver cancer. Indeed, one of the key events involved in HCC progression is excessive levels of reactive oxygen species (ROS) resulting from the fatty acid influx and chronic inflammation. This review provides insights into the different intracellular sources of obesity-induced ROS and molecular mechanisms responsible for hepatic tumorigenesis. In addition, we highlight recent findings pointing to the role of the dysregulated activity of BCL-2 proteins and protein tyrosine phosphatases (PTPs) in the generation of hepatic oxidative stress and ROS-mediated dysfunctional signaling, respectively. Finally, we discuss the potential and challenges of novel nanotechnology strategies to prevent ROS formation in obesity-associated HCC.
Collapse
Affiliation(s)
- Manoja K Brahma
- Signal Transduction and Metabolism Laboratory, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Eduardo H Gilglioni
- Signal Transduction and Metabolism Laboratory, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Lang Zhou
- Materials Research and Education Center, Auburn University, Auburn, AL, 36849, United States
| | - Eric Trépo
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, C.U.B. Hôpital Erasme, Université libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, AL, 36849, United States
| | - Esteban N Gurzov
- Signal Transduction and Metabolism Laboratory, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
36
|
Bello IJ, Oyebode OT, Olanlokun JO, Omodara TO, Olorunsogo OO. Plumbagin induces testicular damage via mitochondrial-dependent cell death. Chem Biol Interact 2021; 347:109582. [PMID: 34302802 DOI: 10.1016/j.cbi.2021.109582] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 06/15/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
Different aspects of reproductive functions are regulated by mitochondrial-controlled events. This study investigated the effect of plumbagin (PL) on testicular mitochondria with a view to unravelling the mechanism of the antifertility potential of plumbagin in testis of healthy rats. Thirty-two male Wistar strain albino rats were randomly allocated into four groups of eight animals each. The control or healthy group received orally 0.1 % DMSO while animals in the remaining three groups received 2.5 mg PL/kg bdwt, 5.0 mg PL/kg bdwt and 10 mg PL/kg bdwt, respectively, for 14 days. In study two, twenty-four male Wistar rats were randomly divided into three (3) groups and were orally administered 0.1% DMSO (control), 30 and 100 mg/kg PL, respectively once daily for 72 h. Rat testis mitochondria were isolated using differential centrifugation. The mitochondrial Permeability Transition (mPT) pore, mitochondrial ATPase (mATPase) activity and mitochondrial lipid peroxidation were assessed spectrophotometrically. Expression of apoptotic proteins (p53, Bax, Bcl-2) and the release of cytochrome c were determined by immunochemical technique. Reproductive receptors (FSH, PR), the expression of aromatase, Testis Specific Kinase-1 {TESK-1} were quantified by RT-PCR. The various doses of plumbagin (2.5, 5.0 and 10 mg/kg bdwt) induced opening of the testicular mPT pore by 2, 5 and 8 folds, respectively, after 14 days of oral administration. These doses of plumbagin also caused enhancement of mATPase activity, elevated generation of mLPO as well as increases in the concentrations of caspases 9 and 3. Sperm analysis revealed that these doses of PL also caused significant decreases in sperm count and motility and increased sperm abnormalities compared to control. Interestingly, these effects were accompanied by dose-dependent expressions of the Bak, p53 and cytochrome c release. Conversely, the abundance of anti-apoptotic Bcl-2 protein decreased relative to control. The levels of transcripts of FSH and progesterone receptors as well as TESK-1 and aromatase decreased significantly relative to control. Furthermore, PL strongly inhibited p53-MDM2 compared to control. Altogether, these findings show that plumbagin damages testicular cells through the activation of mitochondrial pathway involving the p53 protein network.
Collapse
Affiliation(s)
- Isaac J Bello
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olubukola T Oyebode
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - John O Olanlokun
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Todiimu O Omodara
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunso O Olorunsogo
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
37
|
Environmental Factors-Induced Oxidative Stress: Hormonal and Molecular Pathway Disruptions in Hypogonadism and Erectile Dysfunction. Antioxidants (Basel) 2021; 10:antiox10060837. [PMID: 34073826 PMCID: PMC8225220 DOI: 10.3390/antiox10060837] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 01/09/2023] Open
Abstract
Hypogonadism is an endocrine disorder characterized by inadequate serum testosterone production by the Leydig cells of the testis. It is triggered by alterations in the hypothalamic–pituitary–gonadal axis. Erectile dysfunction (ED) is another common disorder in men that involves an alteration in erectile response–organic, relational, or psychological. The incidence of hypogonadism and ED is common in men aged over 40 years. Hypogonadism (including late-onset hypogonadism) and ED may be linked to several environmental factors-induced oxidative stresses. The factors mainly include exposure to pesticides, radiation, air pollution, heavy metals and other endocrine-disrupting chemicals. These environmental risk factors may induce oxidative stress and lead to hormonal dysfunctions. To better understand the subject, the study used many keywords, including “hypogonadism”, “late-onset hypogonadism”, “testosterone”, “erectile dysfunction”, “reactive oxygen species”, “oxidative stress”, and “environmental pollution” in major online databases, such as SCOPUS and PUBMED to extract relevant scientific information. Based on these parameters, this review summarizes a comprehensive insight into the important environmental issues that may have a direct or indirect association with hypogonadism and ED in men. The study concludes that environmental factors-induced oxidative stress may cause infertility in men. The hypothesis and outcomes were reviewed critically, and the mechanistic approaches are applied through oxidant-sensitive pathways. This study also provides reccomendations on future therapeutic interventions and protective measures against such adverse environmental factors-induced hypogonadism and ED.
Collapse
|
38
|
Jia S, Ge S, Fan X, Leong KW, Ruan J. Promoting reactive oxygen species generation: a key strategy in nanosensitizer-mediated radiotherapy. Nanomedicine (Lond) 2021; 16:759-778. [PMID: 33856241 DOI: 10.2217/nnm-2020-0448] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The radiotherapy enhancement effect of numerous nanosensitizers is based on the excessive production of reactive oxygen species (ROS), and only a few systematic reviews have focused on the key strategy in nanosensitizer-mediated radiotherapy. To clarify the mechanism underlying this effect, it is necessary to understand the role of ROS in radiosensitization before clinical application. Thus, the source of ROS and their principle of tumor inhibition are first introduced. Then, nanomaterial-mediated ROS generation in radiotherapy is reviewed. The double-edged sword effect of ROS and the potential dangers they may pose to cancer patients are subsequently addressed. Finally, future perspectives regarding ROS-regulated nanosensitizer applications and development are discussed.
Collapse
Affiliation(s)
- Shichong Jia
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases & Ocular Oncology, Shanghai, 200011, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases & Ocular Oncology, Shanghai, 200011, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases & Ocular Oncology, Shanghai, 200011, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Jing Ruan
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases & Ocular Oncology, Shanghai, 200011, China.,Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| |
Collapse
|
39
|
Wang Y, Qi H, Liu Y, Duan C, Liu X, Xia T, Chen D, Piao HL, Liu HX. The double-edged roles of ROS in cancer prevention and therapy. Theranostics 2021; 11:4839-4857. [PMID: 33754031 PMCID: PMC7978298 DOI: 10.7150/thno.56747] [Citation(s) in RCA: 367] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/31/2021] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen species (ROS) serve as cell signaling molecules generated in oxidative metabolism and are associated with a number of human diseases. The reprogramming of redox metabolism induces abnormal accumulation of ROS in cancer cells. It has been widely accepted that ROS play opposite roles in tumor growth, metastasis and apoptosis according to their different distributions, concentrations and durations in specific subcellular structures. These double-edged roles in cancer progression include the ROS-dependent malignant transformation and the oxidative stress-induced cell death. In this review, we summarize the notable literatures on ROS generation and scavenging, and discuss the related signal transduction networks and corresponding anticancer therapies. There is no doubt that an improved understanding of the sophisticated mechanism of redox biology is imperative to conquer cancer.
Collapse
|
40
|
El Sayed NF, Abdallah DM, Awad AS, Ahmed KA, El-Abhar HS. Novel peripheral role of Nurr-1/GDNF/AKT trajectory in carvedilol and/or morin hydrate hepatoprotective effect in a model of hepatic ischemia/reperfusion. Life Sci 2021; 273:119235. [PMID: 33607152 DOI: 10.1016/j.lfs.2021.119235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/31/2021] [Accepted: 02/08/2021] [Indexed: 12/26/2022]
Abstract
Although the central role of Nurr-1/GDNF has been reviewed amply, scarce data are available on their peripheral impact. Carvedilol and morin hydrate have previously conferred their hepatic anti-fibrotic action. AIM Thus, our aim was to unveil the potential hepatoprotective role of carvedilol (CR) and/or morin hydrate (MH) using a hepatic 70% partial warm ischemia/reperfusion (I/R) rat model. MAIN METHOD Rats were allocated into sham-operated, hepatic I/R, and I/R preceded by oral administration of CR (10 and 30 mg/kg; CR10/CR30), MH (30 mg/kg), or CR10 + MH for one week. KEY FINDINGS On the molecular level, pretreatment with CR and/or MH increased the hepatic contents of Nurr-1, GDNF, and the protein expression of active/p-AKT. On the other hand, they inactivated GSK3β and NF-κB to increase the antioxidant enzymes (GPx, SOD, CAT). All regimens also enhanced the autophagy/lysosomal function and boosted the protein expression of beclin-1, LC3II, and TFEB. Moreover, their antiapoptotic effect was signified by increasing the anti-apoptotic molecule Bcl2 and inhibiting Bax, Bax/Bcl2 ratio, and caspase-3, effects that were confirmed by the TUNEL assay. These improvements were reflected on liver function, as they decreased serum aminotransferases and liver structural alterations induced by I/R. Despite its mild impact, CR10 showed marked improvements when combined with MH; this synergistic interaction overrides the effect of either regimen alone. SIGNIFICANCE In conclusion, CR, MH, and especially the combination regimen, conferred hepatoprotection against I/R via activating the Nurr-1/GDNF/AKT trajectory to induce autophagy/lysosomal biogenesis, inhibit GSK3β/NF-кB hub and apoptosis, and amend redox balance.
Collapse
Affiliation(s)
- Nermein F El Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ahram Canadian University, Cairo, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology &Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Azza S Awad
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ahram Canadian University, Cairo, Egypt
| | - Kawkab A Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt (FUE), 11835 Cairo, Egypt
| |
Collapse
|
41
|
Geniposide, a Principal Component of Gardeniae Fructus, Protects Skin from Diesel Exhaust Particulate Matter-Induced Oxidative Damage. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8847358. [PMID: 33603823 PMCID: PMC7872751 DOI: 10.1155/2021/8847358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 01/05/2021] [Accepted: 01/22/2021] [Indexed: 12/25/2022]
Abstract
Gardeniae Fructus (GF) is the fruit of Gardenia jasminoides Ellis and is traditionally prescribed to treat pyogenic infections and skin ulcers. This study investigated the protective effects of GF and the underlying mechanism responsible for these effects on diesel exhaust particulate matter- (DEP-) induced skin damage. The protective effects of an ethanolic extract of GF (GFE) and its constituents (geniposidic acid, gardenoside, geniposide, chlorogenic acid, and genipin) were examined by analyzing reactive oxygen species (ROS) production, apoptosis, and tight junction (TJ) protein expression in HaCaT cells. Treatment with GFE dose-dependently inhibited intracellular ROS production and apoptosis by regulating the protein expressions of Bax, Bcl-2, and cytochrome C in DEP-stimulated (100 μg/ml) HaCaT cells. Mechanistic studies revealed that the protective effects of GFE were related to its activation of Nrf2 and HO-1 signaling in HaCaT cells. Geniposide, a main constituent of GFE, enhanced the expression of occludin in DEP-stimulated HaCaT cells. Furthermore, topical application of geniposide reduced the expressions of 8-OHdG and Bax and increased the expression of occludin in the dorsal skin lesions of DEP-stimulated mice. Gardeniae Fructus and its main component geniposide are potential candidates for the repair of DEP-induced skin damage due to their antioxidant and antiapoptotic activities.
Collapse
|
42
|
Zhao H, Wang Y, Guo M, Mu M, Yu H, Xing M. Grass carps co-exposed to environmentally relevant concentrations of cypermethrin and sulfamethoxazole bear immunodeficiency and are vulnerable to subsequent Aeromonas hydrophila infection. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 266:115156. [PMID: 32663629 DOI: 10.1016/j.envpol.2020.115156] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 06/11/2023]
Abstract
The aquatic ecosystem is seriously damaged because of the heavy use of pesticides and antibiotics. Fish is the indispensable link between environmental pollution and human health. However, the toxic effects of environment-related concentrations of pesticides and antibiotics in fish have not been thoroughly studied. In this study, grass carps exposed to cypermethrin (CMN, 0.651 μg/L) or/and sulfamethoxazole (SMZ, 0.3 μg/L) for 42 days caused oxidative stress, apoptosis and immunodeficiency in the spleen of grass carps. CMN or/and SMZ exposure led to oxidative damage (consumption of antioxidant enzymes (superoxide dismutase and catalase)) and lipid peroxidation (accumulation of malondialdehyde), induced apoptosis (increases in TUNEL index, Bax/bcl-2, p53, puma and Caspase family expression). In addition, the levels of immunoglobulin M (IgM), complement 3 (C3) were significantly decreased in all treatment groups, which trend was also found in C-reactive protein in CMN and MIX group, and lysozyme in MIX group. Transcription of almost all genes involved in the Toll-like receptors (TLR) signaling pathway was up-regulated under CMN or/and SMZ exposure. However, when subsequently attacked by Aeromonas hydrophila for 2 days, the TLR pathway was inhibited in spleens of all treatment groups accompanied by higher mortality. Overall, the environmentally relevant concentration of CMN and SMZ damages the immune system, triggering oxidative stress and apoptosis in carps. And by affecting the conduction of TLR signaling pathway, CMN or/and SMZ exposure inhibits the innate immune response of fish and reducing their disease resistance. This study highlights the importance of rational and regulated use of these pesticides and antibiotics.
Collapse
Affiliation(s)
- Hongjing Zhao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China; Department of Ecology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China.
| | - Yu Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China.
| | - Menghao Guo
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China
| | - Mengyao Mu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China
| | - Hongxian Yu
- Department of Ecology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China.
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China.
| |
Collapse
|
43
|
Kuriakose GC, Arathi BP, Divya Lakshmanan M, Jiby MV, Gudde RS, Jayabhaskaran C. Sub-acute Toxicity Assessment of Taxol Isolated From Fusarium Solani, an Endophytic Fungus of Taxus Brevifolia, in Wistar Rats and Analyzing Its Cytotoxicity and Apoptotic Potential in Lung Cancer Cells. Front Oncol 2020; 10:538865. [PMID: 33117679 PMCID: PMC7574678 DOI: 10.3389/fonc.2020.538865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 08/14/2020] [Indexed: 01/18/2023] Open
Abstract
The limited availability of taxol from plant sources has prompted the scientific world to look for an alternative, as in the chemical synthesis of tissue cultures of the Taxus species, to meet the increasing demand for the drug. However, these alternative means are expensive or result in low yield. Previously, we have reported that Fusarium solani isolated from Taxus celebica produced taxol and its precursor baccatin III in liquid-grown cultures, and it exhibited promising anticancerous effects in certain cancer cell lines. In the present study, we examined the sub-acute toxicity of fungal taxol (FS) in Wistar rats according to the Organization for Economic Co-operation and Development (OECD) guidelines. The sub-acute oral administration of FS up to 500 mg/kg for a period of 28 days appears to be safe in rats and did not cause severe treatment-related toxicity or treatment-related death. The observed changes in body weight, histopathology, hematological and biochemical parameters, and organ weight were not significant compared to those in the control group of animals. The results suggest that FS is relatively safe when administered orally in rats. The antiproliferative and apoptosis-inducing activities were studied in A549 (human lung cancer) cell line. FS arrested the cells at S and G2/M phases, leading to apoptosis. The characteristic molecular signatures of apoptosis, such as externalized phosphatidyl serine, DNA fragmentation, and nuclear and chromatin condensation, were observed upon FS treatment. FS triggered the generation of reactive oxygen species in A549 cells and elicited cell death by both extrinsic as well as the mitochondria-mediated intrinsic pathway of apoptosis. These results indicate that endophytic fungi isolated from medicinal plants may serve as potential sources of anticancerous compounds with little side effects.
Collapse
Affiliation(s)
- Gini C Kuriakose
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - B P Arathi
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | | | - M V Jiby
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | | | - C Jayabhaskaran
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| |
Collapse
|
44
|
Orabi SH, Allam TS, Shawky SM, Tahoun EAEA, Khalifa HK, Almeer R, Abdel-Daim MM, El-Borai NB, Mousa AA. The Antioxidant, Anti-Apoptotic, and Proliferative Potency of Argan Oil against Betamethasone-Induced Oxidative Renal Damage in Rats. BIOLOGY 2020; 9:E352. [PMID: 33114212 PMCID: PMC7690873 DOI: 10.3390/biology9110352] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/20/2020] [Indexed: 11/16/2022]
Abstract
The present study aimed to investigate the protective effect of argan oil (AO) against nephrotoxic effects following overdose and long-term administration of betamethasone (BM). The phytochemical compositions of AO were assessed using GC/MS. Forty eight male Wister albino rats were divided into six groups and treated for 3 successive weeks. The control group was orally administrated distilled water daily, the BM group received BM (1 mg/kg, IM, day after day), AO/0.5 and AO/1 groups received AO (0.5 mL/kg, 1 mL/kg, orally, daily, respectively), BM + AO/0.5 group and BM + AO/1 group. The results revealed that BM induced hematological changes, including reduction of red blood cells with leukocytosis, neutrophilia, monocytosis, lymphocytopenia, and thrombocytopenia. Moreover, BM caused a significant increase of serum urea and creatinine levels, and renal malondialdehyde and nitric oxide contents with significant decrease of reduced glutathione content. BM also caused vascular, degenerative, and inflammatory histopathological alterations in kidney, along with an increase in the Bax/Bcl-2 ratio, activation of caspase-3, and decrease of proliferating cell nuclear antigen expression. Conversely, the concomitant administration of AO (0.5, 1 mL/kg) with BM ameliorated the aforementioned hematological, biochemical, pathological, and histochemical BM adverse effects. In conclusion, AO has protective effects against BM-induced renal damage, possibly via its antioxidant, anti-apoptotic, and proliferative properties.
Collapse
Affiliation(s)
- Sahar Hassan Orabi
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menofia 32897, Egypt; (H.K.K.); (A.A.M.)
| | - Tamer S. Allam
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menofia 32897, Egypt; or
| | - Sherif Mohamed Shawky
- Department of Physiology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menofia 32897, Egypt;
| | - Enas Abd El-aziz Tahoun
- Department of Pathology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menofia 32897, Egypt;
| | - Hanem K. Khalifa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menofia 32897, Egypt; (H.K.K.); (A.A.M.)
| | - Rafa Almeer
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Nermeen Borai El-Borai
- Department of Forensic Medicine & Toxicology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menofia 32897, Egypt
| | - Ahmed Abdelmoniem Mousa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menofia 32897, Egypt; (H.K.K.); (A.A.M.)
| |
Collapse
|
45
|
Development of Conformational Antibodies to Detect Bcl-xL's Amyloid Aggregates in Metal-Induced Apoptotic Neuroblastoma Cells. Int J Mol Sci 2020; 21:ijms21207625. [PMID: 33076337 PMCID: PMC7589975 DOI: 10.3390/ijms21207625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/31/2022] Open
Abstract
Bcl-xL, a member of the Bcl-2 family, is a pro-survival protein involved in apoptosis regulation. We have previously reported the ability of Bcl-xL to form various types of fibers, from native to amyloid conformations. Here, we have mimicked the effect of apoptosis-induced caspase activity on Bcl-xL by limited proteolysis using trypsin. We show that cleaved Bcl-xL (ΔN-Bcl-xL) forms fibers that exhibit the features of amyloid structures (BclxLcf37). Moreover, three monoclonal antibodies (mAbs), produced by mouse immunization and directed against ΔN-Bcl-xL or Bcl-xL fibers, were selected and characterized. Our results show that these mAbs specifically target ΔN-Bcl-xL in amyloid fibers in vitro. Upon metal-stress-induced apoptosis, these mAbs are able to detect the presence of Bcl-xL in amyloid aggregates in neuroblastoma SH-SY5Y cell lines. In conclusion, these specific mAbs directed against amyloidogenic conformations of Bcl-xL constitute promising tools for studying, in vitro and in cellulo, the contribution of Bcl-xL in apoptosis. These mAbs may further help in developing new diagnostics and therapies, considering Bcl-xL as a strategic target for treating brain lesions relevant to stroke and neurodegenerative diseases.
Collapse
|
46
|
Triterpenoid Saponin AG8 from Ardisia gigantifolia stapf. Induces Triple Negative Breast Cancer Cells Apoptosis through Oxidative Stress Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7963212. [PMID: 33123316 PMCID: PMC7584968 DOI: 10.1155/2020/7963212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/11/2020] [Accepted: 09/12/2020] [Indexed: 11/29/2022]
Abstract
Triple-negative breast cancers (TNBCs) are associated with poor patient survival because of the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expressions. Our previous studies have shown that the triterpenoid saponin AG8 from Ardisia gigantifolia stapf. inhibits the proliferation of MDA-MB-231 cells. In this study, the effects of AG8 were further analyzed in different TNBC cell types: MDA-MB-231, BT-549, and MDA-MB-157 cells. AG8 inhibited the viability of MDA-MB-231, BT-549, and MDA-MB-157 cells in a dose-dependent manner and showed stronger cytotoxicity to African American (AA) and mesenchymal (M) subtypes than Caucasian (CA) and mesenchymal stem-like (MSL) subtypes, respectively. AG8 impaired the uptake of MitoTracker Red CMXRos by the mitochondria of TNBC cells in a dose-dependent manner, and this was recovered by N-acetyl-l-cysteine (NAC). AG8 affected GSH, SOD, and MDA levels of TNBC cells, but different TNBC subtypes had different sensitivities to AG8 and NAC. In addition, we found that AG8 increased the Bax/Bcl-2 ratio and the levels of cytoplasmic cytochrome c and significantly decreased phosphorylation of ERK and AKT in BT549 and MDA-MB-157 cells. AG8 elicited its anticancer effects through ROS generation, ERK and AKT activation, and by triggering mitochondrial apoptotic pathways in TNBC cells. AG8 had selective cytotoxic effects against the AA and M TNBC subtypes and markedly induced MDA-MB-157 (AA subtype) cell apoptosis through pathways that were not associated with ROS, which was different from the other two subtypes. The underlying mechanisms should be further investigated.
Collapse
|
47
|
Do MH, Lee JH, Ahn J, Hong MJ, Kim J, Kim SY. Isosamidin from Peucedanum japonicum Roots Prevents Methylglyoxal-Induced Glucotoxicity in Human Umbilical Vein Endothelial Cells via Suppression of ROS-Mediated Bax/Bcl-2. Antioxidants (Basel) 2020; 9:antiox9060531. [PMID: 32560521 PMCID: PMC7346203 DOI: 10.3390/antiox9060531] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
Methylglyoxal (MGO) is a highly reactive metabolite of glucose. Elevated levels of MGO induce the generation of reactive oxygen species (ROS) and cause cell death in endothelial cells. Vascular endothelial cell damage by ROS has been implicated in the progression of diabetic vascular complications, cardiovascular diseases, and atherosclerosis. In this study, the protective effect of isosamidin, isolated from Peucedanum japonicum roots, on MGO-induced apoptosis was investigated using human umbilical vein endothelial cells (HUVECs). Among the 20 compounds isolated from P. japonicum, isosamidin showed the highest effectiveness in inhibiting MGO-induced apoptosis of HUVECs. Pretreatment of HUVECs with isosamidin significantly prevented the generation of ROS and cell death induced by MGO. Isosamidin prevented MGO-induced apoptosis in HUVECs by downregulating the expression of Bax and upregulating the expression of Bcl-2. MGO treatment activated mitogen-activated protein kinases (MAPKs), such as p38, c-Jun N terminal kinase (JNK), and extracellular signal-regulated kinase (ERK). In contrast, pretreatment with isosamidin strongly inhibited the activation of p38 and JNK. Furthermore, isosamidin caused the breakdown of the crosslinks of the MGO-derived advanced glycation end products (AGEs). These findings suggest that isosamidin from P. japonicum may be used as a preventive agent against MGO-mediated endothelial dysfunction in diabetes. However, further study of the therapeutic potential of isosamidin on endothelial dysfunction needs to explored in vivo models.
Collapse
Affiliation(s)
- Moon Ho Do
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Korea;
| | - Jae Hyuk Lee
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea;
| | - Jongmin Ahn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (J.A.); (M.J.H.); (J.K.)
| | - Min Jee Hong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (J.A.); (M.J.H.); (J.K.)
| | - Jinwoong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (J.A.); (M.J.H.); (J.K.)
| | - Sun Yeou Kim
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea;
- Gachon Institute of Pharmaceutical Science, Gachon University; #191, Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Korea
- Gachon Medical Research Institute, Gil Medical Center, Inchon 21565, Korea
- Correspondence: ; Tel.: +82-32-820-4931; Fax: +82-32-820-4829
| |
Collapse
|
48
|
Zhang X, Cheng J, He P, Zhu J, Chen Z, Miao S, Wang G, Jiang J, Wang Y. Active Monomer RTR-1 Derived from the Root of Rhodomyrtus t omentosa Induces Apoptosis in Gastric Carcinoma Cells by Inducing ER Stress and Inhibiting the STAT3 Signaling Pathway. Cancer Manag Res 2020; 12:3117-3129. [PMID: 32440210 PMCID: PMC7211317 DOI: 10.2147/cmar.s237201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/06/2020] [Indexed: 01/06/2023] Open
Abstract
Purpose Rhodomyrtus tomentosa, a flowering plant from the Myrtaceae family, is considered an antitumour substance with versatile biological and pharmacological activities. RTR-1 is an active monomer purified from the root of Rhodomyrtus tomentosa. However, the detail of mechanism involving in RTR-1 anti-cancer activity remains to be elucidated, and the effect on gastric cancer cells is unknown. Methods Cell proliferation was determined by MTT and clone formation assay. The effect of RTR-1 on cell cycle distribution and apoptosis was analysed utilizing flow cytometry, respectively. Moreover, Western blotting was used to detect the expression of cell cycle- and apoptosis-related protein. Results Based on MTT and clone formation assay, we noticed that RTR-1 inhibited the proliferation of gastric carcinoma (BGC823 and SGC7901) cells in a dose- and time-dependent manner. Furthermore, the results of flow cytometry and Western blotting showed that RTR-1 induced cell cycle arrest in the G2/M phase through the ATM-Chk2-p53-p21 signaling pathway and induced cell apoptosis by inhibiting the signal transducers and activators of transcription 3 (STAT3) pathway and activating the endoplasmic reticulum stress (ER stress) pathway. Conclusion Taken together, these results demonstrate that RTR-1 induces cell cycle arrest and promotes apoptosis in gastric carcinoma, indicating its potential application for gastric cancer therapy.
Collapse
Affiliation(s)
- Xiangqiang Zhang
- Department of Physiology, Basic Medical College, Jinan University, Guangzhou 510630, People's Republic of China
| | - Jinxia Cheng
- Department of Physiology, Basic Medical College, Jinan University, Guangzhou 510630, People's Republic of China
| | - Peiyan He
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou 510630, People's Republic of China
| | - Jinyan Zhu
- Department of Immunology, Basic Medical College, Jinan University, Guangzhou 510630, People's Republic of China
| | - Zhixian Chen
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou 510630, People's Republic of China
| | - Shenyu Miao
- School of Life Sciences, Guangzhou University, Guangzhou, People's Republic of China
| | - Guocai Wang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510630, People's Republic of China
| | - Jianwei Jiang
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou 510630, People's Republic of China
| | - Yuechun Wang
- Department of Physiology, Basic Medical College, Jinan University, Guangzhou 510630, People's Republic of China
| |
Collapse
|
49
|
Wang SW, Lee CH, Lin MS, Chi CW, Chen YJ, Wang GS, Liao KW, Chiu LP, Wu SH, Huang DM, Chen L, Shen YS. ZnO Nanoparticles Induced Caspase-Dependent Apoptosis in Gingival Squamous Cell Carcinoma through Mitochondrial Dysfunction and p70S6K Signaling Pathway. Int J Mol Sci 2020; 21:ijms21051612. [PMID: 32111101 PMCID: PMC7084801 DOI: 10.3390/ijms21051612] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/07/2020] [Accepted: 02/20/2020] [Indexed: 12/17/2022] Open
Abstract
Zinc oxide nanoparticles (ZnO-NPs) are increasingly used in sunscreens, food additives, pigments, rubber manufacture, and electronic materials. Several studies have shown that ZnO-NPs inhibit cell growth and induce apoptosis by the production of oxidative stress in a variety of human cancer cells. However, the anti-cancer property and molecular mechanism of ZnO-NPs in human gingival squamous cell carcinoma (GSCC) are not fully understood. In this study, we found that ZnO-NPs induced growth inhibition of GSCC (Ca9-22 and OECM-1 cells), but no damage in human normal keratinocytes (HaCaT cells) and gingival fibroblasts (HGF-1 cells). ZnO-NPs caused apoptotic cell death of GSCC in a concentration-dependent manner by the quantitative assessment of oligonucleosomal DNA fragmentation. Flow cytometric analysis of cell cycle progression revealed that sub-G1 phase accumulation was dramatically induced by ZnO-NPs. In addition, ZnO-NPs increased the intracellular reactive oxygen species and specifically superoxide levels, and also decreased the mitochondrial membrane potential. ZnO-NPs further activated apoptotic cell death via the caspase cascades. Importantly, anti-oxidant and caspase inhibitor clearly prevented ZnO-NP-induced cell death, indicating the fact that superoxide-induced mitochondrial dysfunction is associated with the ZnO-NP-mediated caspase-dependent apoptosis in human GSCC. Moreover, ZnO-NPs significantly inhibited the phosphorylation of ribosomal protein S6 kinase (p70S6K kinase). In a corollary in vivo study, our results demonstrated that ZnO-NPs possessed an anti-cancer effect in a zebrafish xenograft model. Collectively, these results suggest that ZnO-NPs induce apoptosis through the mitochondrial oxidative damage and p70S6K signaling pathway in human GSCC. The present study may provide an experimental basis for ZnO-NPs to be considered as a promising novel anti-tumor agent for the treatment of gingival cancer.
Collapse
Affiliation(s)
- Shih-Wei Wang
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan;
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Hsing Lee
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ming-Shen Lin
- Department of Water Resources and Environmental Engineering, Tamkang University, New Taipei City 251, Taiwan;
| | - Chih-Wen Chi
- Department of Nursing, MacKay Medical College, New Taipei City 252, Taiwan;
- Department of Medical Research, MacKay Memorial Hospital, Taipei 104, Taiwan
| | - Yu-Jen Chen
- MacKay Junior College of Medicine, Nursing, and Management, Taipei 112, Taiwan;
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei 104, Taiwan
| | - Guo-Shou Wang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan; (G.-S.W.); (K.-W.L.)
- Department of Orthopaedics, MacKay Memorial Hospital, Taipei 104, Taiwan
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan; (G.-S.W.); (K.-W.L.)
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Li-Pin Chiu
- General Education Center, University of Taipei, Taipei 100, Taiwan;
- Division of General Surgery, Taipei City Hospital, Taipei 103, Taiwan
| | - Shu-Hui Wu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 350, Taiwan;
| | - Dong-Ming Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 350, Taiwan;
- Correspondence: (D.-M.H.); (L.C.); (Y.-S.S.); Tel.: +886-37-246-166 (ext. 38105) (D.-M.H.); +886-2-2621-5656 (ext. 2682) (L.C.); +886-2-2636-0303 (ext. 1422) (Y.-S.S.)
| | - Luke Chen
- Department of Water Resources and Environmental Engineering, Tamkang University, New Taipei City 251, Taiwan;
- Correspondence: (D.-M.H.); (L.C.); (Y.-S.S.); Tel.: +886-37-246-166 (ext. 38105) (D.-M.H.); +886-2-2621-5656 (ext. 2682) (L.C.); +886-2-2636-0303 (ext. 1422) (Y.-S.S.)
| | - Yung-Shuen Shen
- Institute of Geriatric Welfare Technology and Science, MacKay Medical College, New Taipei City 252, Taiwan
- Correspondence: (D.-M.H.); (L.C.); (Y.-S.S.); Tel.: +886-37-246-166 (ext. 38105) (D.-M.H.); +886-2-2621-5656 (ext. 2682) (L.C.); +886-2-2636-0303 (ext. 1422) (Y.-S.S.)
| |
Collapse
|
50
|
Perillo B, Di Donato M, Pezone A, Di Zazzo E, Giovannelli P, Galasso G, Castoria G, Migliaccio A. ROS in cancer therapy: the bright side of the moon. Exp Mol Med 2020; 52:192-203. [PMID: 32060354 PMCID: PMC7062874 DOI: 10.1038/s12276-020-0384-2] [Citation(s) in RCA: 1300] [Impact Index Per Article: 260.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) constitute a group of highly reactive molecules that have evolved as regulators of important signaling pathways. It is now well accepted that moderate levels of ROS are required for several cellular functions, including gene expression. The production of ROS is elevated in tumor cells as a consequence of increased metabolic rate, gene mutation and relative hypoxia, and excess ROS are quenched by increased antioxidant enzymatic and nonenzymatic pathways in the same cells. Moderate increases of ROS contribute to several pathologic conditions, among which are tumor promotion and progression, as they are involved in different signaling pathways and induce DNA mutation. However, ROS are also able to trigger programmed cell death (PCD). Our review will emphasize the molecular mechanisms useful for the development of therapeutic strategies that are based on modulating ROS levels to treat cancer. Specifically, we will report on the growing data that highlight the role of ROS generated by different metabolic pathways as Trojan horses to eliminate cancer cells. Highly reactive molecules called reactive oxygen species (ROS), which at low levels are natural regulators of important signaling pathways in cells, might be recruited to act as “Trojan horses” to kill cancer cells. Researchers in Italy led by Bruno Perillo of the Institute of Food Sciences in Avelllino review the growing evidence suggesting that stimulating production of natural ROS species could become useful in treating cancer. Although ROS production is elevated in cancer cells it can also promote a natural process called programmed cell death. This normally regulates cell turnover, but could be selectively activated to target diseased cells. The authors discuss molecular mechanisms underlying the potential anti-cancer activity of various ROS-producing strategies, including drugs and light-stimulated therapies. They expect modifying the production of ROS to have potential for developing new treatments.
Collapse
Affiliation(s)
- Bruno Perillo
- Istituto di Scienze dell'Alimentazione, C.N.R., 83100, Avellino, Italy. .,Istituto per l'Endocrinologia e l'Oncologia Sperimentale, C.N.R., 80131, Naples, Italy.
| | - Marzia Di Donato
- Dipartimento di Medicina di Precisione, Università della Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Antonio Pezone
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Naples, Italy
| | - Erika Di Zazzo
- Dipartimento di Medicina di Precisione, Università della Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Pia Giovannelli
- Dipartimento di Medicina di Precisione, Università della Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Giovanni Galasso
- Dipartimento di Medicina di Precisione, Università della Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Gabriella Castoria
- Dipartimento di Medicina di Precisione, Università della Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Antimo Migliaccio
- Dipartimento di Medicina di Precisione, Università della Campania "L. Vanvitelli", 80138, Naples, Italy
| |
Collapse
|