1
|
Ye J, Sun X, Jiang Q, Gui J, Feng S, Qin B, Xie L, Guo A, Dong J, Sang M. Umbilical cord blood-derived exosomes attenuate dopaminergic neuron damage of Parkinson's disease mouse model. J Nanobiotechnology 2024; 22:567. [PMID: 39277761 PMCID: PMC11401276 DOI: 10.1186/s12951-024-02773-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/14/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Umbilical cord blood (UCB) is a rich source of multifunctional stem cells characterized by low immunogenicity. Recent research in the fields of aging and regenerative medicine has revealed the potential of human umbilical cord blood-derived exosomes (UCB-Exos) in promoting wound healing, anti-aging, and regeneration. However, their role in neurodegenerative diseases, specifically Parkinson's disease (PD), remains unexplored. This study investigates the potential therapeutic effects and underlying mechanisms of UCB-Exos on PD. METHODS Large extracellular vesicles (LEv), Exos, and soluble fractions (SF) of human UCB plasma were extracted to investigate their effects on motor dysfunction of the MPTP-induced PD mouse model and identify the key components that improve PD symptoms. UCB-Exos were administered by the caudal vein to prevent or treat the PD mouse model. The motor function and pathological markers were detected. Differentially expressed gene and KEGG enrichment pathways were screened by transcriptome sequence. MN9D and SH-SY5Y cells were cultured and evaluated for cell viability, oxidative stress, cell cycle, and aging-related indexes by qRT-PCR, western blot, immunofluorescence, and flow cytometry. The protein expression level of the MAPK p38 and ERK1/2 signaling pathway was detected by western blot. RESULTS We observed that LEv, Exos, and SF all exhibited potential in ameliorating motor dysfunction in MPTP-induced PD model mice, with UCB-Exos demonstrating the most significant effect. UCB-Exos showed comparable efficacy in preventing and treating motor dysfunction, cognitive decline, and substantia nigra pathological damage in PD mice. Further investigations revealed that UCB-Exos could potentially alleviate oxidative damage, aging and degeneration, and energy metabolism disorders in neurons. Transcriptome sequencing results corroborated that genes differentially expressed due to UCB-Exos were primarily enriched in the neuroactive ligand-receptor interaction, Dopaminergic synapse, and MAPK signaling pathway. We also observed that UCB-Exos significantly inhibited the hyperphosphorylation of the MAPK p38 and ERK1/2 signaling pathways both in vitro and in vivo. CONCLUSIONS Our study provides a comprehensive evaluation of UCB-Exos on the neuroprotective effects and suggests that inhibition of hyperphosphorylation of MAPK p38 and ERK 1/2 signaling pathways by regulating transcription levels of HspB1 and Ppef2 may be the key mechanism for UCB-Exos to improve PD-related pathological features.
Collapse
Affiliation(s)
- Junjie Ye
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China
- Clinical Laboratory, Wuhan Asia Heart Hospital, Wuhan, 430022, China
| | - Xiaodong Sun
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China
- Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Qi Jiang
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China
| | - Jianjun Gui
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China
| | - Shenglan Feng
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China
| | - Bingqing Qin
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China
| | - Lixia Xie
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China
| | - Ai Guo
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China
| | - Jinju Dong
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China.
| | - Ming Sang
- Research Center for Translational Medicine, Department of Anesthesiology, Department of Obstetrics, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei Provincial Clinical Research Center for Parkinson's Disease at Xiangyang No.1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, China.
- Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| |
Collapse
|
2
|
Huang X, Luodan A, Gao H, He J, Ge L, Cha Z, Gong H, Lin X, Li H, Tang Y, Jiang D, Fan X, Xu H. Mitochondrial transfer between BMSCs and Müller promotes mitochondrial fusion and suppresses gliosis in degenerative retina. iScience 2024; 27:110309. [PMID: 39055937 PMCID: PMC11269791 DOI: 10.1016/j.isci.2024.110309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/19/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Mitochondrial dysfunction and Müller cells gliosis are significant pathological characteristics of retinal degeneration (RD) and causing blinding. Stem cell therapy is a promising treatment for RD, the recently accepted therapeutic mechanism is cell fusion induced materials transfer. However, whether materials including mitochondrial transfer between grafted stem cells and recipient's cells contribute to suppressing gliosis and mechanism are unclear. In present study, we demonstrated that bone marrow mesenchymal stem cells (BMSCs) transferred mitochondria to Müller cells by cell fusion and tunneling nanotubes. BMSCs-derived mitochondria (BMSCs-mito) were integrated into mitochondrial network of Müller cells, improving mitochondrial function, reducing oxidative stress and gliosis, which protected visual function partially in the degenerative rat retina. RNA sequencing analysis revealed that BMSCs-mito increased mitochondrial DNA (mtDNA) content and facilitated mitochondrial fusion in damaged Müller cells. It suggests that mitochondrial transfer from BMSCs remodels Müller cells metabolism and suppresses gliosis; thus, delaying the degenerative progression of RD.
Collapse
Affiliation(s)
- Xiaona Huang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Luodan A
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Hui Gao
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Juncai He
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Lingling Ge
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Zhe Cha
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Hong Gong
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xi Lin
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Huiting Li
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Yongping Tang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Dan Jiang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| |
Collapse
|
3
|
Ezhilarasan D, Ali D, Varghese R. Sesamol induces cytotoxicity via mitochondrial apoptosis in SCC-25 cells. Hum Exp Toxicol 2021; 40:S423-S433. [PMID: 34586880 DOI: 10.1177/09603271211047926] [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] [Indexed: 12/18/2022]
Abstract
Sesamol is the main constituent of sesame seed oil and is obtained from Sesamum indicum. Oral squamous cell carcinoma (OSCC) is one of the most common neoplasms affecting the oral cavity. In this study, we investigated the cytotoxic potentials of sesamol on human oral squamous carcinoma (SCC-25) cells. Human oral squamous carcinoma cells were treated with different concentrations (62.5, 125, and 250 μM/mL) of sesamol for 24 h. Cytotoxicity was analyzed by 3- (4, 5- dimethylthiazol -2- yl) -2, 5-diphenyltetrazolium bromide (MTT) assay. Intracellular reactive oxygen species (ROS) expression was investigated by dichloro-dihydro-fluorescein diacetate assay. Apoptosis-related morphology was analyzed by acridine orange/ethidium bromide staining. Caspase-9 expression was analyzed by confocal microscopic double immunofluorescence staining. Mitochondrial apoptosis-related markers are analyzed using qPCR. Sesamol treatment caused a significant cytotoxic effect in OSCC cells. Sesamol-induced cytotoxic effect was associated with intracellular ROS generation. Sesamol treatments induced a significant increase in the early and late apoptotic cells. This treatment also induced caspase-9 expression in OSCC cells. Sesamol treatments caused downregulation of Harvey rat sarcoma viral oncogene homolog (HRAS) expression at protein and gene levels. Sesamol treatment modulates intrinsic apoptotic marker gene expression in OSCC cells. Overall results confirm the anti-cancer potential of sesamol and it seems to be a promising candidate for OSCC.
Collapse
Affiliation(s)
- D Ezhilarasan
- Department of Pharmacology, The Blue Laboratory, Molecular Medicine and Toxicology Division, Saveetha Dental College and Hospitals, 194347Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - D Ali
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - R Varghese
- Department of Microbiology, Faculty of Science and Informatics, 37442University of Szeged, Szeged, Hungary
| |
Collapse
|
4
|
Karthikkeyan G, Pervaje R, Pervaje SK, Prasad TSK, Modi PK. Prevention of MEK-ERK-1/2 hyper-activation underlines the neuroprotective effect of Glycyrrhiza glabra L. (Yashtimadhu) against rotenone-induced cellular and molecular aberrations. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114025. [PMID: 33775804 DOI: 10.1016/j.jep.2021.114025] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 12/07/2020] [Accepted: 03/10/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yashtimadhu choorna (powder) is prepared from the dried root of Glycyrrhiza glabra L., commonly known as licorice. The Indian Ayurvedic system classifies Yashtimadhu as a Medhya Rasayana that can enhance brain function, improves memory, and possess neuroprotective functions, which can be used against neurodegenerative diseases like Parkinson's disease (PD). AIM OF THE STUDY We aimed to decipher the neuroprotective effects of G. glabra L., i.e., Yashtimadhu, in a rotenone-induced PD model. MATERIALS AND METHODS Retinoic acid-differentiated IMR-32 cells were treated with rotenone (PD model) and Yashtimadhu, and were assessed for cellular toxicity, live-dead staining, cell cycle, oxidative stress, protein abundance, and kinase phosphorylation. RESULTS Yashtimadhu conferred protection against rotenone-induced cytotoxicity, countered cell death, reduced expression of pro-apoptotic proteins (cleaved-caspases-9, and 3, cleaved-PARP, BAX, and BAK) and increased anti-apoptotic protein, BCL-2. Rotenone-induced cell cycle re-entry (G2/M transition), was negated by Yashtimadhu and was confirmed with PCNA levels. Yashtimadhu countered rotenone-mediated activation of mitochondrial proteins involved in oxidative stress, cytochrome-C, PDHA1, and HSP60. Inhibition of rotenone-induced ERK-1/2 hyperphosphorylation prevented activation of apoptosis, which was confirmed with MEK-inhibitor, highlighted the action of Yashtimadhu via ERK-1/2 modulation. CONCLUSIONS We provide the evidence for neuroprotection conferred by G. glabra L. (Yashtimadhu) and its mechanism via inhibiting MEK-ERK-1/2 hyper-phosphorylation, prevention of mitochondrial stress, and subsequent prevention of apoptosis. The study highlights Yashtimadhu as a promising candidate with neuroprotective effects, the potential of which can be harnessed for identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Gayathree Karthikkeyan
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | | | - Sameera Krishna Pervaje
- Yenepoya Medical College and Hospital, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | | | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| |
Collapse
|
5
|
Quinn PMJ, Moreira PI, Ambrósio AF, Alves CH. PINK1/PARKIN signalling in neurodegeneration and neuroinflammation. Acta Neuropathol Commun 2020; 8:189. [PMID: 33168089 PMCID: PMC7654589 DOI: 10.1186/s40478-020-01062-w] [Citation(s) in RCA: 274] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/17/2020] [Indexed: 12/13/2022] Open
Abstract
Mutations in the PTEN-induced kinase 1 (PINK1) and Parkin RBR E3 ubiquitin-protein ligase (PARKIN) genes are associated with familial forms of Parkinson’s disease (PD). PINK1, a protein kinase, and PARKIN, an E3 ubiquitin ligase, control the specific elimination of dysfunctional or superfluous mitochondria, thus fine-tuning mitochondrial network and preserving energy metabolism. PINK1 regulates PARKIN translocation in impaired mitochondria and drives their removal via selective autophagy, a process known as mitophagy. As knowledge obtained using different PINK1 and PARKIN transgenic animal models is being gathered, growing evidence supports the contribution of mitophagy impairment to several human pathologies, including PD and Alzheimer’s diseases (AD). Therefore, therapeutic interventions aiming to modulate PINK1/PARKIN signalling might have the potential to treat these diseases. In this review, we will start by discussing how the interplay of PINK1 and PARKIN signalling helps mediate mitochondrial physiology. We will continue by debating the role of mitochondrial dysfunction in disorders such as amyotrophic lateral sclerosis, Alzheimer’s, Huntington’s and Parkinson’s diseases, as well as eye diseases such as age-related macular degeneration and glaucoma, and the causative factors leading to PINK1/PARKIN-mediated neurodegeneration and neuroinflammation. Finally, we will discuss PINK1/PARKIN gene augmentation possibilities with a particular focus on AD, PD and glaucoma.
Collapse
|
6
|
Chen Y, Hou Y, Ge R, Han J, Xu J, Chen J, Wang H. Protective effect of roscovitine against rotenone-induced parkinsonism. Restor Neurol Neurosci 2018; 36:629-638. [PMID: 30056439 DOI: 10.3233/rnn-180817] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Protective effect of roscovitine and deregulation of the p-RB/E2F1 have not been well studied in PD models generated by repeated oral administration of rotenone. OBJECTIVE These experiments evaluated the effects of repeated oral gavage of rotenone on the activation of p-RB/E2F1 and the effects of roscovitine on the regulation of dopaminergic neuronal injury and the behavior of PD in mice. METHODS Using 2.5% carboxymethylcellulose and 1.25% chloroform as a vehicle solution, rotenone (30 mg/kg) was administered via oral gavage once daily for 30 days in C57 mice. Behavioral profiles (pole test and traction test) were assessed in these PD models, and oxidative stress levels were evaluated in the midbrain. The immunoreactivities of TH, α-synuclein (α-syn), p-RB, E2F1 and cleaved caspase-3 in the substantia nigra were examined with a laser confocal microscope. Pharmacological inhibition of cyclin-dependent kinase with roscovitine was achieved by intraperitoneal (IP) injection at a dose of 50 mg/kg daily. RESULTS All rotenone-administered C57 mice showed the typical behavioral features of PD: stiffness, bradykinesia, or hypokinesia. Behavioral testing with the pole test and traction test indicated that the rotenone group, but not the vehicle group, was affected. Spectrophotometric analysis demonstrated that glutathione (GSH) and superoxide dismutase (SOD) activity was decreased, and the generation of malondialdehyde (MDA) was elevated in the midbrain of the rotenone-treated group. After oral administration of rotenone, a loss of nigral tyrosine hydroxylase (TH)-positive neurons was observed. The immune response of α-syn was enhanced in the cytoplasm of dopaminergic neurons from the rotenone-induced neurotoxicity. Rb phosphorylation at serine 780, which affected Rb binding to E2F, was induced after rotenone treatment. The activation of E2F1, which is involved in the regulation of the cell cycle, was also induced from chronic exposure to rotenone. Moreover, administration of the cell cycle inhibitor roscovitine protected against rotenone-induced nigral dopaminergic neuronal injury and inhibited cleaved caspase-3 activation. Roscovitine also markedly ameliorated the behavior of PD mice. CONCLUSIONS Mouse models of Parkinson's disease were established by oral rotenone administration and reproduced some of the features of dopaminergic neuronal degeneration. Roscovitine protects against rotenone-induced parkinsonism.
Collapse
Affiliation(s)
- Yan Chen
- Department of Gastroenterology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Yiwei Hou
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Ruli Ge
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Jianmei Han
- Department of Neurology, Yangxin County People's Hospital, Shandong Province, China
| | - Jing Xu
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Jinbo Chen
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Hongcai Wang
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| |
Collapse
|
7
|
Abstract
DNA polymerase-β (DNA pol-β) plays a crucial role in the pathogenesis of Parkinson's disease (PD). The aim of this study was to investigate the neuroprotective effects of a DNA polymerase-β inhibitor 2',3'-dideoxycytidine (DDC) in PD models. In the in vitro studies, primary cultured neurons were challenged with 1-methyl-4-phenylpyridinium ion (MPP+). The expression of DNA pol-β was assessed using western blot. The neuroprotective effect of DNA pol-β knockdown and DNA pol-β inhibitor DDC was determined using cell viability assay and caspase-3 activity assay. We found that MPP+ induced neuronal death and the activation of caspase-3 in a dose-dependent manner. The expression of DNA pol-β increased after the neurons were exposed to MPP+. DNA pol-β siRNA or DNA pol-β inhibitor DDC attenuated neuronal death induced by MPP+. In the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD, MPTP treatment triggered behavioral deficits and nigrostriatal lesions. Pretreatment with DDC attenuated MPTP-induced behavioral deficits, dopaminergic neuronal death and striatal dopamine depletion in the MPTP mouse model. These results indicate that DNA pol-β inhibitors may present a novel promising therapeutic option for the neuroprotective treatment of PD.
Collapse
|
8
|
Mustufa MA, Ozen C, Hashmi IA, Aslam A, Baig JA, Yildiz G, Muhammad S, Solangi IB, Ul Hasan Naqvi N, Ozturk M, Ali FI. Synthesis and bio-molecular study of (+)-N-Acetyl-α-amino acid dehydroabietylamine derivative for the selective therapy of hepatocellular carcinoma. BMC Cancer 2016; 16:883. [PMID: 27842576 PMCID: PMC5109647 DOI: 10.1186/s12885-016-2942-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 11/02/2016] [Indexed: 12/13/2022] Open
Abstract
Background The purpose of present work is to synthesize novel (+)-Dehydroabietylamine derivatives (DAAD) using N-acetyl-α-amino acid conjugates and determine its cytotoxic effects on hepatocellular carcinoma cells. Methods An analytical study was conducted to explore cytotoxic activity of DAAD on hepatocellular carcinoma cell lines. The cytotoxicity effect was recorded using sulforhodamine B technique. Cell cycle analysis was performed using Propidium Iodide (PI) staining. Based on cell morphology, anti growth activity and microarray findings of DAAD2 treatment, Comet assay, Annexin V/PI staining, Immunoperoxidase assay and western blots were performed accoringly. Results Hep3B cells were found to be the most sensitive with IC50 of 2.00 ± 0.4 μM against (+)-N-(N-Acetyl-L-Cysteine)-dehydroabietylamine as DAAD2. In compliance to time dependent morphological changes of low cellular confluence, detachment and rounding of DAAD2 treated cells; noticeable changes in G2/M phase were recorded may be leading to cell cycle cessation. Up-regulation (5folds) of TUBA1A gene in Hep3B cells was determined in microarray experiments. Apoptotic mode of cell death was evaluated using standardized staining procedures including comet assay and annexin V/PI staining, Immuno-peroxidase assay. Using western blotting technique, caspase dependant apoptotic mode of cell death was recorded against Hep3B cell line. Conclusion It is concluded that a novel DAAD2 with IC50 values less than 8 μM can induce massive cell attenuation following caspase dependent apoptotic cell death in Hep3B cells. Moreover, the corelation study indicated that DAAD2 may have vital influence on cell prolifration properties. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2942-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Muhammad Ayaz Mustufa
- 5th Floor, PHRC Specialized Research Centre on Child Health, National Institute of Child Health, Karachi, 75500, Pakistan. .,Baqai Institute of Pharmaceutical Sciences (BIPS), Baqai Medical University, Karachi, 74600, Pakistan. .,Department of Molecular, Biology and Genetics, BilGen Genetics and Biotechnology Center, Bilkent University, Ankara, 06800, Turkey.
| | - Cigdem Ozen
- Izmir International Biomedicine and Genome Institute, iBG-izmir, Dokuz Eylül University, 35340, Balcova, Izmir, Turkey
| | - Imran Ali Hashmi
- Department of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Afshan Aslam
- Department of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Jameel Ahmed Baig
- National Center of Excellence in Analytical Chemistry, University of Sindh, Jamshoro, 76080, Pakistan
| | - Gokhan Yildiz
- Department of Medical Biology, Erzincan University Faculty of Medicine, Erzincan, 24100, Turkey
| | - Shoaib Muhammad
- Department of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Imam Bakhsh Solangi
- Dr. M. A. Kazi Institute of Chemistry, University of Sindh, Jamshoro, 76080, Pakistan
| | - Naim Ul Hasan Naqvi
- Baqai Institute of Pharmaceutical Sciences (BIPS), Baqai Medical University, Karachi, 74600, Pakistan
| | - Mehmet Ozturk
- Izmir International Biomedicine and Genome Institute, iBG-izmir, Dokuz Eylül University, 35340, Balcova, Izmir, Turkey
| | - Firdous Imran Ali
- Department of Chemistry, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
9
|
Segura-Aguilar J, Kostrzewa RM. Neurotoxin mechanisms and processes relevant to Parkinson's disease: an update. Neurotox Res 2015; 27:328-54. [PMID: 25631236 DOI: 10.1007/s12640-015-9519-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 01/13/2015] [Accepted: 01/13/2015] [Indexed: 12/14/2022]
Abstract
The molecular mechanism responsible for degenerative process in the nigrostriatal dopaminergic system in Parkinson's disease (PD) remains unknown. One major advance in this field has been the discovery of several genes associated to familial PD, including alpha synuclein, parkin, LRRK2, etc., thereby providing important insight toward basic research approaches. There is an consensus in neurodegenerative research that mitochon dria dysfunction, protein degradation dysfunction, aggregation of alpha synuclein to neurotoxic oligomers, oxidative and endoplasmic reticulum stress, and neuroinflammation are involved in degeneration of the neuromelanin-containing dopaminergic neurons that are lost in the disease. An update of the mechanisms relating to neurotoxins that are used to produce preclinical models of Parkinson´s disease is presented. 6-Hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, and rotenone have been the most wisely used neurotoxins to delve into mechanisms involved in the loss of dopaminergic neurons containing neuromelanin. Neurotoxins generated from dopamine oxidation during neuromelanin formation are likewise reviewed, as this pathway replicates neurotoxin-induced cellular oxidative stress, inactivation of key proteins related to mitochondria and protein degradation dysfunction, and formation of neurotoxic aggregates of alpha synuclein. This survey of neurotoxin modeling-highlighting newer technologies and implicating a variety of processes and pathways related to mechanisms attending PD-is focused on research studies from 2012 to 2014.
Collapse
Affiliation(s)
- Juan Segura-Aguilar
- Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Independencia 1027, Casilla, 70000, Santiago 7, Chile,
| | | |
Collapse
|
10
|
Chiu CC, Yeh TH, Lai SC, Wu-Chou YH, Chen CH, Mochly-Rosen D, Huang YC, Chen YJ, Chen CL, Chang YM, Wang HL, Lu CS. Neuroprotective effects of aldehyde dehydrogenase 2 activation in rotenone-induced cellular and animal models of parkinsonism. Exp Neurol 2014; 263:244-53. [PMID: 25263579 PMCID: PMC4415848 DOI: 10.1016/j.expneurol.2014.09.016] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 09/09/2014] [Accepted: 09/16/2014] [Indexed: 12/17/2022]
Abstract
Many studies have shown that mitochondrial aldehyde dehydrogenase 2 (ALDH2) functions as a cellular protector against oxidative stress by detoxification of cytotoxic aldehydes. Within dopaminergic neurons, dopamine is metabolized by monoamine oxidase to yield 3,4-dihydroxyphenylacetaldehyde (DOPAL) then converts to a less toxic acid product by ALDH. The highly toxic and reactive DOPAL has been hypothesized to contribute to the selective neurodegeneration in Parkinson’s disease (PD). In this study, we investigated the neuroprotective mechanism and therapeutic effect of ALDH2 in rotenone models for parkinsonism. Overexpression of wild-type ALDH2 gene, but not the enzymatically deficient mutant ALDH2*2 (E504K), reduced rotenone-induced cell death. Application of a potent activator of ALDH2, Alda-1, was effective in protecting against rotenone-induced apoptotic cell death in both SH-SY5Y cells and primary cultured substantia nigra (SN) dopaminergic neurons. In addition, intraperitoneal administration of Alda-1 significantly reduced rotenone- or MPTP-induced death of SN tyrosine hydroxylase (TH)-positive dopaminergic neurons. The attenuation of rotenone-induced apoptosis by Alda-1 resulted from decreasing ROS accumulation, reversal of mitochondrial membrane potential depolarization, and inhibition of activation of proteins related to mitochondrial apoptotic pathway. The present study demonstrates that ALDH2 plays a crucial role in maintaining normal mitochondrial function to protect against neurotoxicity and that Alda-1 is effective in ameliorating mitochondrial dysfunction and inhibiting mitochondria-mediated apoptotic pathway. These results indicate that ALDH2 activation could be a neuroprotective therapy for PD.
Collapse
Affiliation(s)
- Ching-Chi Chiu
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Tu-Hsueh Yeh
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Section of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Healthy Aging Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Szu-Chia Lai
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Section of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Healthy Aging Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yah-Huei Wu-Chou
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Human Molecular Genetics Laboratory, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yin-Cheng Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Yu-Jie Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Chao-Lang Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Ya-Ming Chang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Li Wang
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Healthy Aging Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Chin-Song Lu
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Section of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Healthy Aging Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|