1
|
Cheng J, Dong Y, Wu J, Shao C, Tang YC, Dong ZQ. RNA-seq revealed the protective effect of Huangqi Guizhi Wuwu Decoction against cisplatin induced PC12 cell injury. Int J Neurosci 2024:1-11. [PMID: 39155776 DOI: 10.1080/00207454.2024.2392123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy not only affects the tolerability of chemotherapy, but also causes intolerable and prolonged neuropathic pain in cancer patients. Currently, duloxetine is the only drug used to treat chemotherapy-induced peripheral neuropathy. However, the clinical use of this drug still faces several challenges. Therefore, we focused on traditional Chinese medicine to find an effective and safe alternative medicine. Huangqi Guizhi Wuwu Decoction is a traditional Chinese medicine that has been clinically used for treating nerve pain for thousands of years. This study aimed to investigate the neuroprotective effect of Huangqi Guizhi Wuwu Decoction on cisplatin-induced nerve injury in PC12 cells and to elucidate its potential mechanism of action. METHODS Huangqi Guizhi Wuwu Decoction-containing serum and blank serum were prepared from a rat model. The protective effects of Huangqi Guizhi Wuwu Decoction on cisplatin (10 µmol/L)-induced PC12 cell injury were assessed by a Cell Counting Kit-8 assay. RNA expression in Huangqi Guizhi Wuwu Decoction-protected PC12 cells was analyzed using RNA-seq, and subsequently, differentially expressed genes were further analyzed using Gene Ontology and Gene Set Enrichment Analysis. RESULTS The Cell Counting Kit-8 results showed that pretreatment of PC12 cells with Huangqi Guizhi Wuwu Decoction-containing serum (5%, 10%, 15%) significantly increased cells' viability to 10 µmol/L cisplatin-induced cell death. RNA-seq analysis revealed 843 differentially expressed genes in the chemotherapy-induced peripheral neuropathy group and 249 in the Huangqi Guizhi Wuwu Decoction group. The gene set enrichment analysis results in this study suggest that Huangqi Guizhi Wuwu Decoction may treat chemotherapy-induced peripheral neuropathy by enhancing axon guidance. CONCLUSIONS This study provides valuable evidence for using Huangqi Guizhi Wuwu Decoction in treating chemotherapy-induced peripheral neuropathy, partially achieved by improving axon guidance pathways.
Collapse
Affiliation(s)
- Jie Cheng
- Department of Clinical Pharmacy Laboratory, The First Affiliated Hospital of Baotou Medical College, Baotou, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, PR China
| | - Yuan Dong
- Department of Clinical Pharmacy Laboratory, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Jing Wu
- Department of Clinical Pharmacy Laboratory, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Chen Shao
- Department of Clinical Pharmacy Laboratory, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Yu-Chen Tang
- Department of Clinical Pharmacy Laboratory, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Zhi-Qiang Dong
- Department of Clinical Pharmacy Laboratory, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| |
Collapse
|
2
|
Rocha Caldas G, do Amaral L, Munhoz Rodrigues D, Mayrink de Miranda A, Aparecida Guinaim Dos Santos N, Machado Rocha L, Tame Parreira RL, Cardozo Dos Santos A, Kenupp Bastos J. Brazilian Green Propolis' Artepillin C and Its Acetylated Derivative Activate the NGF-Signaling Pathways and Induce Neurite Outgrowth in NGF-Deprived PC12 Cells. Chem Biodivers 2023; 20:e202301294. [PMID: 37953436 DOI: 10.1002/cbdv.202301294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/14/2023]
Abstract
Artepillin C is the most studied compound in Brazilian Green Propolis and, along with its acetylated derivative, displays neurotrophic activity on PC12 cells. Specific inhibitors of the trkA receptor (K252a), PI3K/Akt (LY294002), and MAPK/ERK (U0126) signaling pathways were used to investigate the neurotrophic mechanism. The expression of proteins involved in axonal and synaptic plasticity (GAP-43 and Synapsin I) was assessed by western blotting. Additionally, physicochemical properties, pharmacokinetics, and drug-likeness were evaluated by the SwissADME web tool. Both compounds induced neurite outgrowth by activating the NGF-signaling pathways but through different neuronal proteins. Furthermore, in silico analyses showed interesting physicochemical and pharmacokinetic properties of these compounds. Therefore, these compounds could play an important role in axonal and synaptic plasticity and should be further investigated.
Collapse
Affiliation(s)
- Gabriel Rocha Caldas
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Lilian do Amaral
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Débora Munhoz Rodrigues
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Aline Mayrink de Miranda
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Leandro Machado Rocha
- Natural Products Technology Laboratory-Fluminense Federal University, Niterói, RJ, Brazil
| | | | | | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
3
|
Protective Effect of Natural Antioxidants on Reducing Cisplatin-Induced Nephrotoxicity. DISEASE MARKERS 2022; 2022:1612348. [PMID: 36419843 PMCID: PMC9678481 DOI: 10.1155/2022/1612348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 09/24/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022]
Abstract
The clinical application of cisplatin is limited by its adverse events, of which nephrotoxicity is the most commonly observed. In a cisplatin-induced pathological response, oxidative stress is one of the upstream reactions which inflicts different degrees of damages to the intracellular material components. Reactive oxygen species (ROS) are also one of the early signaling molecules that subsequently undergo a series of pathological reactions, such as apoptosis and necrosis. This review summarizes the mechanism of intracellular ROS generation induced by cisplatin, mainly from the consumption of endogenous antioxidants, destruction of antioxidant enzymes, induction of mitochondrial crosstalk between the endoplasmic reticulum by ROS and Ca2+, and destruction of the cytochrome P450 (CYP) system in the endoplasmic reticulum, all of which result in excessive accumulation of intracellular ROS and oxidative stress. In addition, studies demonstrated that natural antioxidants can protect against the cisplatin-induced nephrotoxicity, by reducing or even eliminating excess free radicals and also affecting other nonredox pathways. Therefore, this review on the one hand provides theoretical support for the research and clinical application of natural antioxidants and on the other hand provides a new entry point for the detailed mechanism of cisplatin nephrotoxicity, which may lay a solid foundation for the future clinical use of cisplatin.
Collapse
|
4
|
Interferon-beta induces major histocompatibility complex of class I (MHC-I) expression and a proinflammatory phenotype in cultivated human astrocytes. Differentiation 2022; 128:43-56. [DOI: 10.1016/j.diff.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/21/2022]
|
5
|
Neuron Compatibility and Antioxidant Activity of Barium Titanate and Lithium Niobate Nanoparticles. Int J Mol Sci 2022; 23:ijms23031761. [PMID: 35163681 PMCID: PMC8836423 DOI: 10.3390/ijms23031761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 12/14/2022] Open
Abstract
The biocompatibility and the antioxidant activity of barium titanate (BaTiO3) and lithium niobate (LiNbO3) were investigated on a neuronal cell line, the PC12, to explore the possibility of using piezoelectric nanoparticles in the treatment of inner ear diseases, avoiding damage to neurons, the most delicate and sensitive human cells. The cytocompatibility of the compounds was verified by analysing cell viability, cell morphology, apoptotic markers, oxidative stress and neurite outgrowth. The results showed that BaTiO3 and LiNbO3 nanoparticles do not affect the viability, morphological features, cytochrome c distribution and production of reactive oxygen species (ROS) by PC12 cells, and stimulate neurite branching. These data suggest the biocompatibility of BaTiO3 and LiNbO3 nanoparticles, and that they could be suitable candidates to improve the efficiency of new implantable hearing devices without damaging the neuronal cells.
Collapse
|
6
|
Sim KH, Lee YJ. Perfluorohexane sulfonate induces memory impairment and downregulation of neuroproteins via NMDA receptor-mediated PKC-ERK/AMPK signaling pathway. CHEMOSPHERE 2022; 288:132503. [PMID: 34626661 DOI: 10.1016/j.chemosphere.2021.132503] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/02/2021] [Accepted: 10/05/2021] [Indexed: 06/13/2023]
Abstract
Perfluorohexane sulfonate (PFHxS) is a widely used industrial chemical detected in human umbilical cord blood and breast milk, and has been suggested to exhibit developmental neurotoxicity. Previous studies on mice reported that neonatal exposure to PFHxS altered neuroprotein levels in the developing brain, and caused behavioral toxicity and cognitive dysfunction in the mature brain. However, the underlying mechanisms responsible for PFHxS-induced neuroprotein dysregulation are poorly understood. In this study, we examined the effect of neonatal exposure to PFHxS on memory function using an in vivo mice model. Furthermore, we examined the levels of growth associated protein-43 (GAP-43) and calcium/calmodulin dependent protein kinase II (CaMKII) (biomarkers of neuronal development) and the involved signaling pathways using differentiated neuronal PC12 cells. PFHxS decreased cell viability, GAP-43 and CaMKII levels, and neurite formation. These effects were mediated by the NMDA receptor, PKC-α, PKC-δ, AMPK and ERK pathways. MK801, an NMDA receptor antagonist, reduced the activation of PKC-α, PKC-δ, ERK and AMPK. The activation of ERK was suppressed by pharmacological and knockdown inhibition of PKC-α and -δ. Interestingly, the AMPK pathway was selectively inhibited by inhibiting PKC-δ but not PKC-ɑ. Consistent with PFHxS-induced neuronal death, and GAP-43 and CaMKII downregulation, neonatal exposure to PFHxS caused significant memory impairment in adult mice. Collectively, these results demonstrate that PFHxS induces persistent developmental neurotoxicity, as well as GAP-43 and CaMKII downregulation via the NMDA receptor-mediated PKCs (α and δ)-ERK/AMPK pathways.
Collapse
Affiliation(s)
- Kyeong Hwa Sim
- Department of Pharmacology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Youn Ju Lee
- Department of Pharmacology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea.
| |
Collapse
|
7
|
A Review of the Health Protective Effects of Phenolic Acids against a Range of Severe Pathologic Conditions (Including Coronavirus-Based Infections). Molecules 2021; 26:molecules26175405. [PMID: 34500838 PMCID: PMC8433690 DOI: 10.3390/molecules26175405] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/18/2022] Open
Abstract
Phenolic acids comprise a class of phytochemical compounds that can be extracted from various plant sources and are well known for their antioxidant and anti-inflammatory properties. A few of the most common naturally occurring phenolic acids (i.e., caffeic, carnosic, ferulic, gallic, p-coumaric, rosmarinic, vanillic) have been identified as ingredients of edible botanicals (thyme, oregano, rosemary, sage, mint, etc.). Over the last decade, clinical research has focused on a number of in vitro (in human cells) and in vivo (animal) studies aimed at exploring the health protective effects of phenolic acids against the most severe human diseases. In this review paper, the authors first report on the main structural features of phenolic acids, their most important natural sources and their extraction techniques. Subsequently, the main target of this analysis is to provide an overview of the most recent clinical studies on phenolic acids that investigate their health effects against a range of severe pathologic conditions (e.g., cancer, cardiovascular diseases, hepatotoxicity, neurotoxicity, and viral infections—including coronaviruses-based ones).
Collapse
|
8
|
Lv L, Cui H, Ma Z, Liu X, Yang L. Recent progresses in the pharmacological activities of caffeic acid phenethyl ester. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:1327-1339. [PMID: 33492405 DOI: 10.1007/s00210-021-02054-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/14/2021] [Indexed: 12/16/2022]
Abstract
The past decades have seen a growing interest in natural products. Caffeic acid phenethyl ester (CAPE), a flavonoid isolated from honeybee propolis, has shown multiple pharmacological potentials, including anti-cancer, anti-inflammatory, antioxidant, antibacterial, antifungal, and protective effects on nervous systems and multiple organs, since it was found as a potent nuclear factor κB (NF-κB) inhibitor. This review summarizes the advances in these beneficial effects of CAPE, as well as the underlying mechanisms, and proposes that CAPE offers an opportunity for developing therapeutics in multiple diseases. However, clinical trials on CAPE are necessary and encouraged to obtain certain clinically relevant conclusions.
Collapse
Affiliation(s)
- Lili Lv
- Jilin University, Changchun, 130021, China
| | | | - Zhiming Ma
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Xin Liu
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Longfei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
9
|
Şahin A, Kaya S, Baylan M. The effects of caffeic acid phenethyl ester on retina in a diabetic rat model. Cutan Ocul Toxicol 2021; 40:268-273. [PMID: 34165369 DOI: 10.1080/15569527.2021.1940196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE We aimed to investigate the effect of caffeic acid phenethyl ester (CAPE) on retinal apoptosis and oxidative stress parameters in streptozotocin (STZ) induced diabetic rat model. METHODS This study included 3 groups; control, STZ, and STZ + CAPE. The rats in STZ, and STZ + CAPE groups were injected with STZ (35 mg/kg, i.p.) for induction of diabetes. In the STZ + CAPE group, 10 µmol/kg of CAPE were intraperitoneally injected for 4 weeks. Control and STZ groups were given only intraperitoneal vehicle (saline). Rats were anaesthetized and sacrificed on the 4th week of the experiment. Total anti-oxidant status (TAS), and total oxidant status (TOS) were measured on the dissected retinal tissues. Oxidative stress index (OSI) was also calculated. Fellow eyes were used for histopathologic evaluation with caspase-3 and matrix metalloproteinase-2 (MMP-2) and MMP-9 evaluation. RESULTS TAS levels were similar between groups (p = 0.71). However, CAPE treatment prevented the elevation of the TOS in the STZ + CAPE group compared to the STZ group (30.93 ± 9.97 vs 61.53 ± 24.7 nmol H2O2 Eq/mg protein, p = 0.007). OSI was also significantly lower in the STZ + CAPE group than that of the STZ group (20.01 ± 5.87 vs. 37.90 ± 14.32, respectively, p = 0.007). Retinal caspase-3 staining, MMP-2 and MMP-9 scores were not different between groups (p > 0.05 for all). CONCLUSION The present study demonstrated that CAPE treatment may decrease the oxidative stress in the retina in STZ induced diabetic rat model. However, apoptosis was not observed in the retina. The retinal apoptosis cannot be shown probably due to a shorter period of diabetes.
Collapse
Affiliation(s)
- Alparslan Şahin
- Department of Ophthalmology, Memorial Dicle Hospital, Diyarbakır, Turkey
| | - Savaş Kaya
- Department of Immunology, School of Medicine, Dicle University, Diyarbakır, Turkey
| | - Mukadder Baylan
- Department of Physiology, School of Medicine, Dicle University, Diyarbakır, Turkey
| |
Collapse
|
10
|
Liu BW, Zhang J, Hong YS, Li NB, Liu Y, Zhang M, Wu WY, Zheng H, Lampert A, Zhang XW. NGF-Induced Nav1.7 Upregulation Contributes to Chronic Post-surgical Pain by Activating SGK1-Dependent Nedd4-2 Phosphorylation. Mol Neurobiol 2021; 58:964-982. [PMID: 33063281 DOI: 10.1007/s12035-020-02156-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/29/2020] [Indexed: 01/07/2023]
Abstract
At present, chronic post-surgical pain (CPSP) is difficult to prevent and cure clinically because of our lack of understanding of its mechanisms. Surgical injury induces the upregulation of voltage-gated sodium channel Nav1.7 in dorsal root ganglion (DRG) neurons, suggesting that Nav1.7 is involved in the development of CPSP. However, the mechanism leading to persistent dysregulation of Nav1.7 is largely unknown. Given that nerve growth factor (NGF) induces a long-term increase in the neuronal hyperexcitability after injury, we hypothesized that NGF might cause the long-term dysregulation of Nav1.7. In this study, we aimed to investigate whether Nav1.7 regulation by NGF is involved in CPSP and thus contributes to the specific mechanisms involved in the development of CPSP. Using conditional nociceptor-specific Nav1.7 knockout mice, we confirmed the involvement of Nav1.7 in NGF-induced pain and identified its role in the maintenance of pain behavior during long-term observations (up to 14 days). Using western blot analyses and immunostaining, we showed that NGF could trigger the upregulation of Nav1.7 expression and thus support the development of CPSP in rats. Using pharmacological approaches, we showed that the increase of Nav1.7 might be partly regulated by an NGF/TrkA-SGK1-Nedd4-2-mediated pathway. Furthermore, reversing the upregulation of Nav1.7 in DRG could alleviate spinal sensitization. Our results suggest that the maintained upregulation of Nav1.7 triggered by NGF contributes to the development of CPSP. Attenuating the dysregulation of Nav1.7 in peripheral nociceptors may be a strategy to prevent the transition from acute post-surgical pain to CPSP.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animals
- Behavior, Animal/drug effects
- Benzamides/pharmacology
- Brain-Derived Neurotrophic Factor/metabolism
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Hydrazines/pharmacology
- Immediate-Early Proteins/antagonists & inhibitors
- Immediate-Early Proteins/metabolism
- Indoles/pharmacology
- Male
- Mice, Knockout
- Models, Biological
- NAV1.7 Voltage-Gated Sodium Channel/genetics
- NAV1.7 Voltage-Gated Sodium Channel/metabolism
- Nedd4 Ubiquitin Protein Ligases/metabolism
- Nerve Growth Factor/pharmacology
- Pain, Postoperative/genetics
- Pain, Postoperative/pathology
- Phosphorylation/drug effects
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Rats, Sprague-Dawley
- Receptor, trkA/antagonists & inhibitors
- Receptor, trkA/metabolism
- Spinal Cord/pathology
- Ubiquitination/drug effects
- Up-Regulation/drug effects
- Vesicular Glutamate Transport Protein 2/metabolism
- Mice
- Rats
Collapse
Affiliation(s)
- Bao-Wen Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jin Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi-Shun Hong
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ning-Bo Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mi Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen-Yao Wu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hua Zheng
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Angelika Lampert
- Institute of Physiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Xian-Wei Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
11
|
Balaha M, De Filippis B, Cataldi A, di Giacomo V. CAPE and Neuroprotection: A Review. Biomolecules 2021; 11:biom11020176. [PMID: 33525407 PMCID: PMC7911454 DOI: 10.3390/biom11020176] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Propolis, a product of the honey bee, has been used in traditional medicine for many years. A hydrophobic bioactive polyphenolic ester, caffeic acid phenethyl ester (CAPE), is one of the most extensively investigated active components of propolis. Several studies have indicated that CAPE has a broad spectrum of pharmacological activities as anti-oxidant, anti-inflammatory, anti-viral, anti-fungal, anti-proliferative, and anti-neoplastic properties. This review largely describes CAPE neuroprotective effects in many different conditions and summarizes its molecular mechanisms of action. CAPE was found to have a neuroprotective effect on different neurodegenerative disorders. At the basis of these effects, CAPE has the ability to protect neurons from several underlying causes of various human neurologic diseases, such as oxidative stress, apoptosis dysregulation, and brain inflammation. CAPE can also protect the nervous system from some diseases which negatively affect it, such as diabetes, septic shock, and hepatic encephalopathy, while numerous studies have demonstrated the neuroprotective effects of CAPE against adverse reactions induced by different neurotoxic substances. The potential role of CAPE in protecting the central nervous system (CNS) from secondary injury following various CNS ischemic conditions and CAPE anti-cancer activity in CNS is also reviewed. The structure–activity relationship of CAPE synthetic derivatives is discussed as well.
Collapse
Affiliation(s)
- Marwa Balaha
- Department of Pharmacy, University “G. d’Annunzio”, Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (B.D.F.); (V.d.G.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, 33516 Kafr El Sheikh, Egypt
| | - Barbara De Filippis
- Department of Pharmacy, University “G. d’Annunzio”, Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (B.D.F.); (V.d.G.)
| | - Amelia Cataldi
- Department of Pharmacy, University “G. d’Annunzio”, Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (B.D.F.); (V.d.G.)
- Correspondence: ; Tel.: +39-0871-355-4467
| | - Viviana di Giacomo
- Department of Pharmacy, University “G. d’Annunzio”, Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (B.D.F.); (V.d.G.)
| |
Collapse
|
12
|
Ceylan T, Kaymak E, Cantürk Tan F, Yakan B. Research on the protective effect of caffeic acid phenethyl ester on testicular damage caused by cisplatin. Turk J Med Sci 2020; 50:2032-2039. [PMID: 32628437 PMCID: PMC7775695 DOI: 10.3906/sag-2002-58] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/04/2020] [Indexed: 12/18/2022] Open
Abstract
Background/aim Cisplatin (CP), a chemotherapeutic drug, causes damage to spermatogenic serial cells, Sertoli cells, and Leydig cells in rat testicles. It was aimed to investigate the protective effect of caffeic acid phenethyl ester (CAPE), one of the active ingredients of propolis, in eliminating CP-induced testicular damage. Materials and methods Group 1 (control group) was given physiological saline solution intraperitoneally (IP) throughout the experiment. Group 2 (CP group) was given a single dose of CP (7 mg/kg) IP on the day 7. Group 3 (CP + CAPE group), was given CAPE (10 µmol/kg/day) IP for 12 days and a single dose of CP (7 mg/kg) IP on day 7. Group 4 (CAPE group) was given CAPE (10 µmol/kg/day) IP for 12 days. On day 14 of the experiment, the rats were decapitated under xylazine and ketamine anesthesia and their testicles were removed. The sections obtained from the testicles were stained with hematoxylin-eosin and histopathological damage was evaluated. Malondialdehyde (MDA) levels, and superoxide dismutase (SOD) and catalase (CAT) enzymatic activities were measured in the testicular tissue samples. Testosterone (TES) levels were measured in the blood serum. The Johnsen testicular biopsy score (JTBS) was used to evaluate testicular tubules. DNA damage was evaluated in sperm samples taken from the ductus epididymis using the comet assay technique. Results In Group 2, which was given CP, the testicles were severely damaged. It was observed that histological damage was reduced in the testes by administering CAPE in Group 3. Moreover, according to the JTBS, the value was significantly higher in the testicular tubules (P < 0.05). Moreover, the MDA level decreased in Group 3. However, the SOD, CAT, and TES levels increased in Group 3. DNA damage also decreased significantly in Group 3 when compared to Group 2 (P < 0.05). Conclusion The results showed that CAPE may be protective against damage caused by CP in the testicles of rats.
Collapse
Affiliation(s)
- Tayfun Ceylan
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Emin Kaymak
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Fazile Cantürk Tan
- Department of Biophysics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Birkan Yakan
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
13
|
Ali AM, Kunugi H. Apitherapy for Parkinson's Disease: A Focus on the Effects of Propolis and Royal Jelly. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1727142. [PMID: 33123309 PMCID: PMC7586183 DOI: 10.1155/2020/1727142] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/27/2020] [Accepted: 10/05/2020] [Indexed: 02/08/2023]
Abstract
The vast increase of world's aging populations is associated with increased risk of age-related neurodegenerative diseases such as Parkinson's disease (PD). PD is a widespread disorder characterized by progressive loss of dopaminergic neurons in the substantia nigra, which encompasses a wide range of debilitating motor, emotional, cognitive, and physical symptoms. PD threatens the quality of life of millions of patients and their families. Additionally, public welfare and healthcare systems are burdened with its high cost of care. Available treatments provide only a symptomatic relief and produce a trail of noxious side effects, which increase noncompliance. Hence, researchers have recently focused on the use of nutraceuticals as safe adjunctive treatments of PD to limit its progress and associated damages in affected groups. Propolis is a common product of the beehive, which possesses a large number of therapeutic properties. Royal jelly (RJ) is a bee product that is fed to bee queens during their entire life, and it contributes to their high physical fitness, fertility, and long lifespan. Evidence suggests that propolis and RJ can promote health by preventing the occurrence of age-related debilitating diseases. Therefore, they have been used to treat various serious disorders such as diabetes mellitus, cardiovascular diseases, and cancer. Some evolving studies used these bee products to treat PD in animal models. However, a clear understanding of the collective effect of propolis and RJ as well as their mechanism of action in PD is lacking. This review evaluates the available literature for the effects of propolis and RJ on PD. Whenever possible, it elaborates on the underlying mechanisms through which they function in this disorder and offers insights for fruitful use of bee products in future clinical trials.
Collapse
Affiliation(s)
- Amira Mohammed Ali
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Psychiatric Nursing and Mental Health, Faculty of Nursing, Alexandria University, Alexandria, Egypt
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Psychiatry, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Caffeic acid phenethyl ester counteracts doxorubicin-induced chemobrain in Sprague-Dawley rats: Emphasis on the modulation of oxidative stress and neuroinflammation. Neuropharmacology 2020; 181:108334. [PMID: 33011199 DOI: 10.1016/j.neuropharm.2020.108334] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/12/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022]
Abstract
Chemotherapy-induced cognitive dysfunction (chemobrain) is one of the major complaints for cancer patients treated with chemotherapy such as Doxorubicin (DOX). The induction of oxidative stress and neuroinflammation were identified as major contributors to such adverse effect. Caffeic acid phenethyl ester (CAPE) is a natural polyphenolic compound, that exhibits unique context-dependent antioxidant activity. It exhibits pro-oxidant effects in cancer cells, while it is a potent antioxidant and cytoprotective in normal cells. The present study was designed to investigate the potential neuroprotective effects of CAPE against DOX-induced cognitive impairment. Chemobrain was induced in Sprague Dawley rats via systemic DOX administration once per week for 4 weeks (2 mg/kg/week, i.p.). CAPE was administered at 10 or 20 μmol/kg/day, i.p., 5 days per week for 4 weeks. Morris water maze (MWM) and passive avoidance tests were used to assess learning and memory functions. Oxidative stress was evaluated via the colorimetric determination of GSH and MDA levels in both hippocampal and prefrontal cortex brain regions. However, inflammatory markers, acetylcholine levels, and neuronal cell apoptosis were assessed in the same brain areas using immunoassays including either ELISA, western blotting or immunohistochemistry. DOX produced significant impairment in learning and memory as indicated by the data generated from MWM and step-through passive avoidance tests. Additionally DOX-triggered oxidative stress as evidenced from the reduction in GSH levels and increased lipid peroxidation. Treatment with DOX resulted in neuroinflammation as indicated by the increase in NF-kB (p65) nuclear translocation in addition to boosting the levels of pro-inflammatory mediators (COX-II/TNF-α) along with the increased levels of glial fibrillary acid protein (GFAP) in the tested tissues. Moreover, DOX reduced acetylcholine levels and augmented neuronal cell apoptosis as supported by the increased active caspase-3 levels. Co-treatment with CAPE significantly counteracted DOX-induced behavioral and molecular abnormalities in rat brain tissues. Our results provide the first preclinical evidence for CAPE promising neuroprotective activity against DOX-induced neurodegeneration and memory deficits.
Collapse
|
15
|
Evaluation of the neuroprotective potential of caffeic acid phenethyl ester in a cellular model of Parkinson's disease. Eur J Pharmacol 2020; 883:173342. [PMID: 32634439 DOI: 10.1016/j.ejphar.2020.173342] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 01/23/2023]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, and oxidative stress and mitochondrial dysfunction play a major role in the pathogenesis of PD. Since conventional therapeutics are not sufficient for the treatment of PD, the development of new agents with anti-oxidant potential is crucial. Caffeic Acid Phenethyl Ester (CAPE), a biologically active flavonoid of propolis, possesses several biological properties such as immunomodulatory, anti-inflammatory and anti-oxidative. In the present study, we investigated the neuroprotective effects of CAPE against 6-hydroxydopamine (6-OHDA)-induced SH-SY5Y cells. The neuroprotective effects were detected by using cell viability, Annexin V, Hoechst staining, total caspase activity, cell cycle, as well as western blotting. Besides, the anti-oxidative activity was measured by the production of reactive oxygen species and mitochondrial function was determined by measurement of mitochondrial membrane potential (ΔΨm). We found that CAPE significantly increased cell viability and decreased apoptotic cell death (~20%) after 150 μM 6-OHDA exposure following 24 h. 1.25 μM CAPE also prevented 6-OHDA-induced changes in condensed nuclear morphology. Furthermore, treatment with 1.25 μM CAPE increased mitochondrial membrane potential in 6-OHDA-exposed cells. CAPE inhibited 6-OHDA-induced caspase activity (~2 fold) and production of reactive oxygen species. In addition, 150 μM 6-OHDA-induced down-regulation of Bcl-2 and Akt levels and up-regulation of Bax and cleaved caspase-9/caspase-9 levels were partially restored by 1.25 μM CAPE treatment. These results revealed a neuroprotective potential of CAPE against 6-OHDA-induced apoptosis in an in vitro PD model and may be a potential therapeutic candidate for the prevention of neurodegeneration in Parkinson's Disease.
Collapse
|
16
|
Sari AN, Bhargava P, Dhanjal JK, Putri JF, Radhakrishnan N, Shefrin S, Ishida Y, Terao K, Sundar D, Kaul SC, Wadhwa R. Combination of Withaferin-A and CAPE Provides Superior Anticancer Potency: Bioinformatics and Experimental Evidence to Their Molecular Targets and Mechanism of Action. Cancers (Basel) 2020; 12:E1160. [PMID: 32380701 PMCID: PMC7281427 DOI: 10.3390/cancers12051160] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
We have earlier reported anticancer activity in Withaferin A (Wi-A), a withanolide derived from Ashwagandha (Withania somnifera) and caffeic acid phenethyl ester (CAPE), an active compound from New Zealand honeybee propolis. Whereas Wi-A was cytotoxic to both cancer and normal cells, CAPE has been shown to cause selective death of cancer cells. In the present study, we investigated the efficacy of Wi-A, CAPE, and their combination to ovarian and cervical cancer cells. Both Wi-A and CAPE were seen to activate tumor suppressor protein p53 by downregulation of mortalin and abrogation of its interactions with p53. Downregulation of mortalin translated to compromised mitochondria integrity and function that affected poly ADP-ribose polymerase1 (PARP1); a key regulator of DNA repair and protein-target for Olaparib, drugs clinically used for treatment of breast, ovarian and cervical cancers)-mediated DNA repair yielding growth arrest or apoptosis. Furthermore, we also compared the docking capability of Wi-A and CAPE to PARP1 and found that both of these could bind to the catalytic domain of PARP1, similar to Olaparib. We provide experimental evidences that (i) Wi-A and CAPE cause inactivation of PARP1-mediated DNA repair leading to accumulation of DNA damage and activation of apoptosis signaling by multiple ways, and (ii) a combination of Wi-A and CAPE offers selective toxicity and better potency to cancer cells.
Collapse
Affiliation(s)
- Anissa Nofita Sari
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (A.N.S.); (P.B.); (J.K.D.); (J.F.P.)
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Priyanshu Bhargava
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (A.N.S.); (P.B.); (J.K.D.); (J.F.P.)
| | - Jaspreet Kaur Dhanjal
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (A.N.S.); (P.B.); (J.K.D.); (J.F.P.)
- DAILAB, Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi 110 016, India; (N.R.); (S.S.); (D.S.)
| | - Jayarani F. Putri
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (A.N.S.); (P.B.); (J.K.D.); (J.F.P.)
| | - Navaneethan Radhakrishnan
- DAILAB, Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi 110 016, India; (N.R.); (S.S.); (D.S.)
| | - Seyad Shefrin
- DAILAB, Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi 110 016, India; (N.R.); (S.S.); (D.S.)
| | - Yoshiyuki Ishida
- CycloChem Co. Ltd., 7-4-5 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan; (Y.I.); (K.T.)
| | - Keiji Terao
- CycloChem Co. Ltd., 7-4-5 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan; (Y.I.); (K.T.)
| | - Durai Sundar
- DAILAB, Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi 110 016, India; (N.R.); (S.S.); (D.S.)
| | - Sunil C. Kaul
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (A.N.S.); (P.B.); (J.K.D.); (J.F.P.)
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Renu Wadhwa
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (A.N.S.); (P.B.); (J.K.D.); (J.F.P.)
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
17
|
PC12 Cell Line: Cell Types, Coating of Culture Vessels, Differentiation and Other Culture Conditions. Cells 2020; 9:cells9040958. [PMID: 32295099 PMCID: PMC7227003 DOI: 10.3390/cells9040958] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/09/2020] [Accepted: 04/12/2020] [Indexed: 12/27/2022] Open
Abstract
The PC12 cell line is one of the most commonly used in neuroscience research, including studies on neurotoxicity, neuroprotection, neurosecretion, neuroinflammation, and synaptogenesis. Two types of this line are available in the ATCC collection: traditional PC12 cells grown in suspension and well-attached adherent phenotype. PC12 cells grown in suspension tend to aggregate and adhere poorly to non-coated surfaces. Therefore, it is necessary to modify the surface of culture vessels. This paper aims to characterise the use of two distinct variants of PC12 cells as well as describe their differentiation and neuronal outgrowth with diverse NGF concentrations (rat or human origin) on various surfaces. In our study, we evaluated cell morphology, neurite length, density and outgrowth (measured spectrofluorimetrically), and expression of neuronal biomarkers (doublecortin and NeuN). We found that the collagen coating was the most versatile method of surface modification for both cell lines. For adherent cells, the coating was definitely less important, and the poly-d-lysine surface was as good as collagen. We also demonstrated that the concentration of NGF is of great importance for the degree of differentiation of cells. For suspension cells, we achieved the best neuronal characteristics (length and density of neurites) after 14 days of incubation with 100 ng/mL NGF (change every 48 h), while for adherent cells after 3-5 days, after which they began to proliferate. In the PC12 cell line, doublecortin (DCX) expression in the cytoplasm and NeuN in the cell nucleus were found. In turn, in the PC12 Adh line, DCX was not expressed, and NeuN expression was located in the entire cell (both in the nucleus and cytoplasm). Only the traditional PC12 line grown in suspension after differentiation with NGF should be used for neurobiological studies, especially until the role of the NeuN protein, whose expression has also been noted in the cytoplasm of adherent cells, is well understood.
Collapse
|
18
|
Mohd Sairazi NS, Sirajudeen KNS. Natural Products and Their Bioactive Compounds: Neuroprotective Potentials against Neurodegenerative Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:6565396. [PMID: 32148547 PMCID: PMC7042511 DOI: 10.1155/2020/6565396] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/09/2019] [Accepted: 01/06/2020] [Indexed: 02/06/2023]
Abstract
In recent years, natural products, which originate from plants, animals, and fungi, together with their bioactive compounds have been intensively explored and studied for their therapeutic potentials for various diseases such as cardiovascular, diabetes, hypertension, reproductive, cancer, and neurodegenerative diseases. Neurodegenerative diseases, including Alzheimer's disease, Huntington's disease, Parkinson's disease, and amyotrophic lateral sclerosis are characterized by the progressive dysfunction and loss of neuronal structure and function that resulted in the neuronal cell death. Since the multifactorial pathological mechanisms are associated with neurodegeneration, targeting multiple mechanisms of actions and neuroprotection approach, which involves preventing cell death and restoring the function to damaged neurons, could be promising strategies for the prevention and therapeutic of neurodegenerative diseases. Natural products have emerged as potential neuroprotective agents for the treatment of neurodegenerative diseases. This review focused on the therapeutic potential of natural products and their bioactive compounds to exert a neuroprotective effect on the pathologies of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nur Shafika Mohd Sairazi
- Faculty of Medicine, Universiti Sultan Zainal Abidin (UniSZA), Medical Campus, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia
| | - K. N. S. Sirajudeen
- Department of Chemical Pathology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia, Bandar Indera Mahkota, 25200 Kuantan, Pahang, Malaysia
| |
Collapse
|
19
|
Santos NAGD, Ferreira RS, Santos ACD. Overview of cisplatin-induced neurotoxicity and ototoxicity, and the protective agents. Food Chem Toxicol 2019; 136:111079. [PMID: 31891754 DOI: 10.1016/j.fct.2019.111079] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022]
Abstract
Cisplatin has dramatically improved the survival rate of cancer patients, but it has also increased the prevalence of hearing and neurological deficits in this population. Cisplatin induces ototoxicity, peripheral (most prevalent) and central (rare) neurotoxicity. This review addresses the ototoxicity and the neurotoxicity associated with cisplatin-based chemotherapy, providing an integrated view of the potential protective agents that have been evaluated in vitro, in vivo and in clinical trials, their targets and mechanisms of protection and their effects on the antitumor activity of cisplatin. So far, the findings are insufficient to support the use of any oto- or neuroprotective agent before, during or after cisplatin chemotherapy. Despite their promising effects in vitro and in animal studies, many agents have not been evaluated in clinical trials. Additionally, the clinical trials have limitations concerning the sample size, controls, measurement, heterogeneous groups, several arms of treatment, short follow-up or no blinding. Besides that, for most agents, the effects on the antitumor activity of cisplatin have not been evaluated in tumor-bearing animals, which discourages clinical trials. Further well-designed randomized controlled clinical trials are necessary to definitely demonstrate the effectiveness of the oto- or neuroprotective agents proposed by animal and in vitro studies.
Collapse
Affiliation(s)
- Neife Aparecida Guinaim Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaela Scalco Ferreira
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Antonio Cardozo Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
20
|
FA-97, a New Synthetic Caffeic Acid Phenethyl Ester Derivative, Protects against Oxidative Stress-Mediated Neuronal Cell Apoptosis and Scopolamine-Induced Cognitive Impairment by Activating Nrf2/HO-1 Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8239642. [PMID: 31885818 PMCID: PMC6914945 DOI: 10.1155/2019/8239642] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/30/2019] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder with cognitive deficits, which is becoming markedly more common in the world. Currently, the exact cause of AD is still unclear, and no curative therapy is available for preventing or mitigating the disease progression. Caffeic acid phenethyl ester (CAPE), a natural phenolic compound derived from honeybee hive propolis, has been reported as a potential therapeutic agent against AD, while its application is limited due to the low water solubility and poor bioavailability. Here, caffeic acid phenethyl ester 4-O-glucoside (FA-97) is synthesized. We validate that FA-97 attenuates H2O2-induced apoptosis in SH-SY5Y and PC12 cells and suppresses H2O2-induced oxidative stress by inhibiting the ROS level, malondialdehyde (MDA) level, and protein carbonylation level, as well as induces cellular glutathione (GSH) and superoxide dismutase (SOD). Mechanistically, FA-97 promotes the nuclear translocation and transcriptional activity of Nrf2 associated with the upregulated expression of HO-1 and NQO-1. The prime importance of Nrf2 activation in the neuroprotective and antioxidant effects of FA-97 is verified by Nrf2 siRNA transfection. In addition, FA-97 prevents scopolamine- (SCOP-) induced learning and memory impairments in vivo via reducing neuronal apoptosis and protecting against cholinergic system dysfunction in the hippocampus and cortex. Moreover, the increased MDA level and low total antioxidant capacity in SCOP-treated mouse brains are reversed by FA-97, with the increased expression of HO-1, NQO-1, and nuclear Nrf2. In conclusion, FA-97 protects against oxidative stress-mediated neuronal cell apoptosis and SCOP-induced cognitive impairment by activating Nrf2/HO-1 signaling, which might be developed as a therapeutic drug for AD.
Collapse
|
21
|
Ferreira RS, Dos Santos NAG, Bernardes CP, Sisti FM, Amaral L, Fontana ACK, Dos Santos AC. Caffeic Acid Phenethyl Ester (CAPE) Protects PC12 Cells Against Cisplatin-Induced Neurotoxicity by Activating the AMPK/SIRT1, MAPK/Erk, and PI3k/Akt Signaling Pathways. Neurotox Res 2019; 36:175-192. [PMID: 31016689 DOI: 10.1007/s12640-019-00042-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/26/2019] [Accepted: 04/04/2019] [Indexed: 01/01/2023]
Abstract
Peripheral sensory neuropathy (PSN) is a well-known side effect of cisplatin characterized by axonal damage. In the early stage of neurotoxicity, cisplatin affects proteins that modulate neurite outgrowth and neuroplasticity, without inducing mitochondrial damage or apoptosis. There are no preventive therapies for cisplatin-induced peripheral neuropathy; therefore, measures to improve axonal growth and connectivity would be beneficial. Caffeic acid phenethyl ester (CAPE) is a bioactive component of propolis with neurotrophic and neuroprotective activities. We have recently showed that CAPE protects against cisplatin-induced neurotoxicity by activating NGF high-affinity receptors (trkA) and inducing neuroplasticity. We have now assessed other potential early targets of cisplatin and additional mechanisms involved in the neuroprotection of CAPE. Cisplatin reduced axonal cytoskeletal proteins (F-actin and β-III-tubulin) without inducing oxidative damage in PC12 cells. It also reduced energy-related proteins (AMPK α, p-AMPK α, and SIRT1) and glucose uptake. At this stage of neurotoxicity, glutamate excitotoxicity is not involved in the toxicity of cisplatin. CAPE attenuated the downregulation of the cytoskeleton and energy-related markers as well as SIRT1 and phosphorylated AMPK α. Moreover, the neuroprotective mechanism of CAPE also involves the activation of the neurotrophic signaling pathways MAPK/Erk and PI3k/Akt. The PI3K/Akt pathway is involved in the upregulation of SIRT1 induced by CAPE, but not in the upregulation of cytoskeletal proteins. Altogether, these findings suggest that the neuroprotective effect of CAPE against cisplatin-induced neurotoxicity involves both (a) a neurotrophic mechanism that mimics the mechanism triggered by the NGF itself and (b) a non-neurotrophic mechanism that upregulates the cytoskeletal proteins.
Collapse
Affiliation(s)
- Rafaela Scalco Ferreira
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Neife Aparecida Guinaim Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Carolina P Bernardes
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Flávia Malvestio Sisti
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Lilian Amaral
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Andreia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Antonio Cardozo Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
22
|
Donertas B, Unel CC, Erol K. Cannabinoids and agmatine as potential therapeutic alternatives for cisplatin-induced peripheral neuropathy. J Exp Pharmacol 2018; 10:19-28. [PMID: 29950907 PMCID: PMC6018893 DOI: 10.2147/jep.s162059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cisplatin is a widely used antineoplastic agent in the treatment of various cancers. Peripheral neuropathy is a well-known side effect of cisplatin and has the potential to result in limiting and/or reducing the dose, decreasing the quality of life. Unfortunately, the mechanism for cisplatin-induced neuropathy has not been completely elucidated. Currently, available treatments for neuropathic pain (NP) are mostly symptomatic, insufficient and are often linked with several detrimental side effects; thus, effective treatments are needed. Cannabinoids and agmatine are endogenous modulators that are implicated in painful states. This review explains the cisplatin-induced neuropathy and antinociceptive effects of cannabinoids and agmatine in animal models of NP and their putative therapeutic potential in cisplatin-induced neuropathy.
Collapse
Affiliation(s)
- Basak Donertas
- Department of Medical Pharmacology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Cigdem Cengelli Unel
- Department of Medical Pharmacology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Kevser Erol
- Department of Medical Pharmacology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
23
|
Mazzio E, Badisa R, Eyunni S, Ablordeppey S, George B, Soliman KFA. Bioactivity-Guided Isolation of Neuritogenic Factor from the Seeds of the Gac Plant ( Momordica cochinchinensis). EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:8953958. [PMID: 29955238 PMCID: PMC6000838 DOI: 10.1155/2018/8953958] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/02/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022]
Abstract
Nerve growth factor (NGF) is an endogenously produced protein with the capacity to induce central nervous system (CNS) neuronal differentiation and repair. NGF signaling involves its binding to tropomyosin-related kinase (Trk) receptors, internalization, and initiation of phosphorylation cascades which cause microtubule reorganization and neuronal outgrowth. Because NGF cannot cross the blood-brain barrier, its therapeutic use is limited. Synthetic peptides that can act as NGF receptor agonists (NGF mimetics) are known to attenuate neurodegenerative pathologies in experimental models of Alzheimer's disease and Parkinson's disease; however, the existence of plant-based NGF mimetics is uncertain. For this reason, we recently completed a high throughput screening of over 1100 nutraceuticals (vitamins, herbal plant parts, polyphenolics, teas, fruits, and vegetables) to identify neuritogenic factor using a PC-12 cell model. Remarkably we found only one, commonly known as the seed of Gac plant (Momordica cochinchinensis) (MCS). In the current study, we further investigated this seed for its neuritogenic effect using bioactivity-guided chemical separations. The data show no biological neuritogenic activity in any chemical solvent fraction, where activity was exclusive to the crude protein. MSC crude proteins were then separated by 1D electrophoresis, where the active neuritogenic activity was confirmed to have a molecular mass of approximately 17 kDa. Subsequently, the 17kDa band was excised, digested, and run on a UPLC-MS/MS with a Q Exactive Hybrid Quadrupole-Orbitrap Mass Spectrometer with data evaluated diverse tools such as X! Tandem, OMS, and K-score algorithms. Proteomic evaluation of the 17kDa band confirmed evidence for 11S globulin subunit beta, napin, oleosin, Momordica trypsin inhibitors (TI) MCoTI-I /II, and many isoforms of Two Inhibitor Peptide Topologies (TIPTOPs). While all peptides identified correspond to the genus/species, Momordica cochinchinensis and Cucumis Sativus, a significant limitation of the analysis is the nonexistence of full annotation for the Momordica cochinchinensis proteome. In conclusion, these findings demonstrate that there is a stable protein within MCS having a mass of 17kDa with the capacity to induce neurite outgrowth. Future work will be required to establish the therapeutic value of the MCS for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- E. Mazzio
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - R. Badisa
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - S. Eyunni
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - S. Ablordeppey
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - B. George
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - K. F. A. Soliman
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|