1
|
Tsai KW, Liao JB, Tseng HW. Metformin regulates the proliferation and motility of melanoma cells by modulating the LINC00094/miR-1270 axis. Cancer Cell Int 2024; 24:384. [PMID: 39563323 PMCID: PMC11575040 DOI: 10.1186/s12935-024-03545-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Melanoma is an aggressive tumor with a high mortality rate. Metformin, a commonly prescribed diabetes medication, has shown promise in cancer prevention and treatment. Long noncoding RNAs (lncRNAs) are non-protein-coding RNA molecules that play a key role in tumor development by interacting with cellular chromatins. Despite the benefits of metformin, the anticancer mechanism underlying its effect on the regulation of lncRNAs in melanoma remains unclear. METHODS We investigated the lncRNA profiles of human melanoma cells with and without metformin treatment using a next-generation sequencing approach (NGS). Utilizing public databases, we analyzed the expression levels and clinical impacts of LINC00094 and miR-1270 in melanoma. The expression levels of LINC00094 and miR-1270 were verified in human cell lines and clinical samples by real-time PCR and in situ hybridization. The biological roles of LINC00094 and miR-1270 in cell growth, proliferation, cell cycle, apoptosis, and motility were studied using in vitro assays. RESULTS We identify a novel long noncoding RNA, namely LINC00094, whose expression considerably decreased in melanoma cells after metformin treatment. In situ hybridization analysis revealed substantially higher expression of LINC00094 in cutaneous melanoma tissue compared with adjacent normal epidermis and normal control tissues (P < 0.001). In nondiabetic patients with melanoma, the overall survival of high LINC00094 expression group was shorter than the low LINC00094 expression group with borderline statistical significance (log-rank test, P = 0.057). Coexpression analysis of LINC00094 indicated its involvement in the mitochondrial respiratory pathway, with its knockdown suppressing genes associated with mitochondrial oxidative phosphorylation, glycolysis, antioxidant production, and metabolite levels. Functional analysis revealed that silencing-LINC00094 inhibited the proliferation, colony formation, invasion, and migration of melanoma cells. Cell cycle analysis following LINC00094 knockdown revealed G1 phase arrest with reduced cell cycle protein expression. Combined TargetScan and reporter assays revealed a direct link between miR-1270 and LINC00094. Ectopic miR-1270 expression inhibited melanoma cell growth and motility while inducing apoptosis. Finally, through in silico analysis, we identified two miR-1270 target genes, CD276 and centromere protein M (CENPM), which may be involved in the biological functions of LINC00094. CONCLUSIONS Overall, LINC00094 expression may regulate melanoma cell growth and motility by modulating the expression of miR-1270, and targeting genes of CD276 and CENPM indicating its therapeutic potential in melanoma treatment.
Collapse
Affiliation(s)
- Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, New Taipei City, Taiwan
| | - Jia-Bin Liao
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Shu Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Hui-Wen Tseng
- Department of Dermatology, Ministry of Health and Welfare Pingtung Hospital, Pingtung, Taiwan.
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan.
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan.
- Department of Nursing, College of Nursing, Meiho University, Neipu, Pingtung, Taiwan.
| |
Collapse
|
2
|
Yang L, Zhang Y, Hua J, Song G, Li F, Zheng N, Zhang T, Xu Z, Ren X, Zhu B, Han Y, Guo Y, Han J, Zhou B. Integrated Studies on Male Reproductive Toxicity of Decabromodiphenyl Ethane in Zebrafish Spermatozoa Ex Vivo, Male Zebrafish in Vivo, and GC-1 Cells in Vitro. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:117005. [PMID: 39570742 PMCID: PMC11580837 DOI: 10.1289/ehp14426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 09/06/2024] [Accepted: 10/24/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Legacy brominated flame retardants have been recognized as risky factors leading to declined sperm quality. The widespread utilization of decabromodiphenyl ethane (DBDPE) as a replacement for decabromodiphenyl ether has given rise to considerable concern over its potential risks to reproductive health. OBJECTIVES The objectives were to quickly determine whether DBDPE affects sperm quality upon ex vivo exposure, to reveal the reproductive outcomes and underlying molecular mechanisms using an in vivo zebrafish model exposed to DBDPE, and to validate the potential impact on DNA damage and energy metabolism balance in vitro. METHODS Zebrafish spermatozoa were treated with DBDPE (0.01, 0.1, 1, 10 μ M ) for 3 h, and the spermatozoa motility and fertilization ability with normal eggs were evaluated. Then adult male zebrafish were treated with DBDPE (0.1, 1, 10, and 100 nM ) for 2 months, and their reproductive performance was examined. Four-dimensional label-free proteome and phosphoproteome were performed in zebrafish testes, and the findings were validated by multiple indicators. Finally, mouse spermatogonial GC-1 cells were treated with DBDPE (0.1, 1 μ M ) for 72 h, and DNA damage was examined, as well as the energy production of glycolysis and oxidative phosphorylation. RESULTS Ex vivo exposure to DBDPE caused lower motility and fertilization rates of zebrafish spermatozoa. In vivo exposure to DBDPE caused lower sperm motility and abnormal spermatogenesis in male zebrafish testes. Integrated whole-proteome and phosphoproteome analysis revealed DNA damage responses and energy metabolic disorders in zebrafish testes. A dosage window characterized by higher mitochondrial membrane potential (MMP) in combination with unchanged reactive oxygen species and apoptosis rates was observed in both zebrafish testes and GC-1 cells. DISCUSSION This study suggests that in zebrafish, DBDPE exposure could impair sperm quality and spermatogenesis, and the underlying mechanism could be related to DNA damage and energy metabolic reprogramming in testicular germ cells. https://doi.org/10.1289/EHP14426.
Collapse
Affiliation(s)
- Lihua Yang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Yindan Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Jianghuan Hua
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
- Hubei Shizhen Laboratory, Wuhan, People’s Republic of China
| | - Guili Song
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Fan Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Na Zheng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Taotao Zhang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Zhixiang Xu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, People’s Republic of China
| | - Xinxin Ren
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Biran Zhu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Yanna Han
- Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resource and Environmental Engineering, East China University of Science and Technology, Shanghai, People’s Republic of China
| | - Yongyong Guo
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Jian Han
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Bingsheng Zhou
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| |
Collapse
|
3
|
Zhao X, Wang F, Kam C, Wu MY, Zhang J, Xu C, Bao K, He Q, Ye R, Tang BZ, Chen S. Fluorescent Nanocable as a Biomedical Tool: Intracellular Self-Assembly Formed by a Natural Product Interconnects and Synchronizes Mitochondria. ACS NANO 2024; 18:21447-21458. [PMID: 39080909 PMCID: PMC11328177 DOI: 10.1021/acsnano.4c06186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 08/18/2024]
Abstract
Self-assembly processes commonly occur in various biological contexts to form functional biological structures. However, the self-assembly of nanofibers within cells by heterologous molecules showing a biological function is rare. In this work, we reported the intracellular formation of fluorescent nanofibers by a natural small molecule, lycobetaine (LBT), which facilitated the direct physical connection between mitochondria and synchronized their membrane potential oscillations. The luminescent properties of LBT enabled the real-time observation of nanofiber formation, while the semiconductive nature of the LBT nanofiber facilitated electrical signal transduction among the connected mitochondria. This study introduces an approach to modulate mitochondrial connectivity within cells using "nano-cables" which facilitate studies on synchronized mitochondrial operations and the underlying mechanisms of drug action.
Collapse
Affiliation(s)
- Xueqian Zhao
- School
of Life Sciences, The Chinese University
of Hong Kong, Hong Kong 999077, China
- Ming
Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong 999077, China
| | - Fei Wang
- School
of Life Sciences, The Chinese University
of Hong Kong, Hong Kong 999077, China
- Ming
Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong 999077, China
| | - Chuen Kam
- School
of Life Sciences, The Chinese University
of Hong Kong, Hong Kong 999077, China
- Ming
Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong 999077, China
| | - Ming-Yu Wu
- Ming
Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong 999077, China
| | - Jianyu Zhang
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, The Hong Kong University of Science
and Technology, Hong Kong 999077, China
| | - Changhuo Xu
- Ministry
of Education Frontiers Science Center for Precision Oncology, Faculty
of Health Sciences, University of Macau, Macao 999078, China
| | - Kai Bao
- Department
of Materials Science and Engineering, Department of Chemistry, City University of Hong Kong, Hong Kong 999077, China
| | - Qiyuan He
- Department
of Materials Science and Engineering, Department of Chemistry, City University of Hong Kong, Hong Kong 999077, China
| | - Ruquan Ye
- Department
of Materials Science and Engineering, Department of Chemistry, City University of Hong Kong, Hong Kong 999077, China
| | - Ben Zhong Tang
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, The Hong Kong University of Science
and Technology, Hong Kong 999077, China
- School of
Science and Engineering, Shenzhen Institute of Aggregate Science and
Technology, The Chinese University of Hong
Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Sijie Chen
- School
of Life Sciences, The Chinese University
of Hong Kong, Hong Kong 999077, China
- Ming
Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong 999077, China
| |
Collapse
|
4
|
Zheng N, Wang X, Zhang Y, Hua J, Zhu B, Zhou Y, Xu Z, Luo L, Han J, Yang L, Zhou B. Mechanistic Insights into 1,2-bis(2,4,6-tribromophenoxy)ethane-Induced Male Reproductive Toxicity in Zebrafish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:8251-8263. [PMID: 38695612 DOI: 10.1021/acs.est.4c00849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2024]
Abstract
The novel brominated flame retardant, 1,2-bis(2,4,6-tribromophenoxy)ethane (BTBPE), has increasingly been detected in environmental and biota samples. However, limited information is available regarding its toxicity, especially at environmentally relevant concentrations. In the present study, adult male zebrafish were exposed to varying concentrations of BTBPE (0, 0.01, 0.1, 1, and 10 μg/L) for 28 days. The results demonstrated underperformance in mating behavior and reproductive success of male zebrafish when paired with unexposed females. Additionally, a decline in sperm quality was confirmed in BTBPE-exposed male zebrafish, characterized by decreased total motility, decreased progressive motility, and increased morphological malformations. To elucidate the underlying mechanism, an integrated proteomic and phosphoproteomic analysis was performed, revealing a predominant impact on mitochondrial functions at the protein level and a universal response across different cellular compartments at the phosphorylation level. Ultrastructural damage, increased expression of apoptosis-inducing factor, and disordered respiratory chain confirmed the involvement of mitochondrial impairment in zebrafish testes. These findings not only provide valuable insights for future evaluations of the potential risks posed by BTBPE and similar chemicals but also underscore the need for further research into the impact of mitochondrial dysfunction on reproductive health.
Collapse
Affiliation(s)
- Na Zheng
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaochen Wang
- Ecology and Environment Monitoring and Scientific Research Center, Ecology and Environment Administration of Yangtze River Basin, Ministry of Ecology and Environment, Wuhan 430010, China
| | - Yindan Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianghuan Hua
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Biran Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yuxi Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Zhixiang Xu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Lijun Luo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Jian Han
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
5
|
Zhang S, Mei Y, Liu J, Liu Z, Tian Y. Alkyne-tagged SERS nanoprobe for understanding Cu + and Cu 2+ conversion in cuproptosis processes. Nat Commun 2024; 15:3246. [PMID: 38622137 PMCID: PMC11018805 DOI: 10.1038/s41467-024-47549-1] [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: 10/27/2023] [Accepted: 03/29/2024] [Indexed: 04/17/2024] Open
Abstract
Simultaneously quantifying mitochondrial Cu+ and Cu2+ levels is crucial for evaluating the molecular mechanisms of copper accumulation-involved pathological processes. Here, a series of molecules containing various diacetylene derivatives as Raman reporters are designed and synthesized, and the alkyne-tagged SERS probe is created for determination Cu+ and Cu2+ with high selectivity and sensitivity. The developed SERS probe generates well-separated distinguishable Raman fingerprint peaks with built-in corrections in the cellular silent region, resulting in accurate quantification of Cu+ and Cu2+. The present probe demonstrates high tempo-spatial resolution for real-time imaging and simultaneously quantifying mitochondrial Cu+ and Cu2+ with long-term stability benefiting from the probe assembly with designed Au-C≡C groups. Using this powerful tool, it is found that mitochondrial Cu+ and Cu2+ increase during ischemia are associated with breakdown of proteins containing copper as well as conversion of Cu+ and Cu2+. Meanwhile, we observe that parts of Cu+ and Cu2+ are transported out of neurons by ATPase. More importantly, cuproptosis in neurons is found including the oxidative stress process caused by the conversion of Cu+ to Cu2+, which dominates at the early stage (<9 h), and subsequent proteotoxic stress. Both oxidative and proteotoxic stresses contribute to neuronal death.
Collapse
Affiliation(s)
- Sihan Zhang
- State Key Laboratory of Molecular & Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai, China
| | - Yuxiao Mei
- State Key Laboratory of Molecular & Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai, China
| | - Jiaqi Liu
- State Key Laboratory of Molecular & Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai, China
| | - Zhichao Liu
- State Key Laboratory of Molecular & Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai, China.
| | - Yang Tian
- State Key Laboratory of Molecular & Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai, China.
| |
Collapse
|
6
|
Öğüten PN, Engür Öztürk S, Dikmen M. The investigation of cytotoxic and apoptotic activity of Cl-amidine on the human U-87 MG glioma cell line. Medicine (Baltimore) 2024; 103:e37015. [PMID: 38394536 PMCID: PMC11309612 DOI: 10.1097/md.0000000000037015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/02/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Peptidyl (protein) arginine deiminases (PADs) provide the transformation of peptidyl arginine to peptidyl citrulline in the presence of calcium with posttranslational modification. The dysregulated PAD activity plays an important role on too many diseases including also the cancer. In this study, it has been aimed to determine the potential cytotoxic and apoptotic activity of chlorine-amidine (Cl-amidine) which is a PAD inhibitor and whose effectiveness has been shown in vitro and in vivo studies recently on human glioblastoma cell line Uppsala 87 malignant glioma (U-87 MG) forming an in vitro model for the glioblastoma multiforme (GBM) which is the most aggressive and has the highest mortality among the brain tumors. METHODS In the study, the antiproliferative and apoptotic effects of Cl-amidine on GBM cancer model were investigated. The antiproliferative effects of Cl-amidine on U-87 MG cells were determined by 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate method at the 24th and 48th hours. The apoptotic effects were analyzed by Annexin V and Propidium iodide staining, caspase-3 activation, and mitochondrial membrane polarization (5,5', 6,6'-tetrachloro-1,1', 3,3' tetraethyl benzimidazolyl carbocyanine iodide) methods in the flow cytometry. RESULTS It has been determined that Cl-amidine exhibits notable antiproliferative properties on U-87 MG cell line in a time and concentration-dependent manner, as determined through the 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate assay. Assessment of apoptotic effects via Annexin V and Propidium iodide staining and 5,5', 6,6'-tetrachloro-1,1', 3,3' tetraethyl benzimidazolyl carbocyanine iodide methods has revealed significant efficacy, particularly following a 24-hour exposure period. It has been observed that Cl-amidine induces apoptosis in cells by enhancing mitochondrial depolarization, independently of caspase-3 activation. Furthermore, regarding its impact on healthy cells, it has been demonstrated that Cl-amidine shows lower cytotoxic effects when compared to carmustine, an important therapeutic agent for glioblastoma. CONCLUSION The findings of this study have shown that Cl-amidine exhibits significant potential as an anticancer agent in the treatment of GBM. This conclusion is based on its noteworthy antiproliferative and apoptotic effects observed in U-87 MG cells, as well as its reduced cytotoxicity toward healthy cells in comparison to existing treatments. We propose that the antineoplastic properties of Cl-amidine should be further investigated through a broader spectrum of cancer cell types. Moreover, we believe that investigating the synergistic interactions of Cl-amidine with single or combination therapies holds promise for the discovery of novel anticancer agents.
Collapse
Affiliation(s)
- Pinar Naile Öğüten
- Faculty of Medicine, Department of Histology, Samsun University, Samsun, Turkey
| | - Selin Engür Öztürk
- Department of Pharmacy Services, Tavas Vocational School of Health Services, Pamukkale University, Denizli, Turkey
| | - Miriş Dikmen
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
7
|
Algothmi KM, Mahasneh ZMH, Abdelnour SA, Khalaf QAW, Noreldin AE, Barkat RA, Khalifa NE, Khafaga AF, Tellez-Isaias G, Alqhtani AH, Swelum AA, Abd El-Hack ME. Protective impacts of mitochondria enhancers against thermal stress in poultry. Poult Sci 2024; 103:103218. [PMID: 37980733 PMCID: PMC10692709 DOI: 10.1016/j.psj.2023.103218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 11/21/2023] Open
Abstract
Heat stress (HS) is still the essential environmental agent influencing the poultry industry. Research on HS in poultry has progressively acquired growing interest because of increased attention to climate alteration. Poultry can survive at certain zone of environmental temperatures, so it could be considered homoeothermic. In poultry, the normal body temperature is essential to enhance the internal environment for growth, which is achieved by normal environmental temperature. Recently, many studies have revealed that HS could cause mitochondrial dysfunction in broilers by inducing redox dysfunction, increasing uncoupling protein, boosting lipid and protein oxidation, and oxidative stress. Moreover, HS diminished the energy suppliers supported by mitochondria activity. A novel strategy for combating the negative influences of HS via boosting the mitochondria function through enrichment of the diets with mitochondria enhancers was also described in this review. Finally, the current review highlights the mitochondria dysfunction induced by HS in broilers and attempts to boost mitochondria functionality by enriching mitochondria enhancers to broiler diets.
Collapse
Affiliation(s)
- Khloud M Algothmi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Zeinab M H Mahasneh
- Department of Animal Production, School of Agriculture, the University of Jordan, Amman 11942, Jordan
| | - Sameh A Abdelnour
- Department of Animal Production, Faculty of Agriculture, Zagazig University, Zagazig 44519, Egypt
| | - Qahtan A W Khalaf
- Department of Medical Laboratory Techniques, College of Medical Technology, Al-Kitab University, Kirkuk 36001, Iraq
| | - Ahmed E Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Rasha A Barkat
- Department of Physiology, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Norhan E Khalifa
- Department of Physiology, Faculty of Veterinary Medicine, Matrouh University, Matrouh 51744, Egypt
| | - Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria 22758, Egypt
| | | | - Abdulmohsen H Alqhtani
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Ayman A Swelum
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mohamed E Abd El-Hack
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt.
| |
Collapse
|
8
|
Miller ZA, Mueller A, Kim T, Jolivert JF, Ma RZ, Muthuswami S, Park A, McMahon DB, Nead KT, Carey RM, Lee RJ. Lidocaine induces apoptosis in head and neck squamous cell carcinoma through activation of bitter taste receptor T2R14. Cell Rep 2023; 42:113437. [PMID: 37995679 PMCID: PMC10842818 DOI: 10.1016/j.celrep.2023.113437] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/22/2023] [Accepted: 10/29/2023] [Indexed: 11/25/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) have high mortality and significant treatment-related morbidity. It is vital to discover effective, minimally invasive therapies that improve survival and quality of life. Bitter taste receptors (T2Rs) are expressed in HNSCCs, and T2R activation can induce apoptosis. Lidocaine is a local anesthetic that also activates bitter taste receptor 14 (T2R14). Lidocaine has some anti-cancer effects, but the mechanisms are unclear. Here, we find that lidocaine causes intracellular Ca2+ mobilization through activation of T2R14 in HNSCC cells. T2R14 activation with lidocaine depolarizes mitochondria, inhibits proliferation, and induces apoptosis. Concomitant with mitochondrial Ca2+ influx, ROS production causes T2R14-dependent accumulation of poly-ubiquitinated proteins, suggesting that proteasome inhibition contributes to T2R14-induced apoptosis. Lidocaine may have therapeutic potential in HNSCCs as a topical gel or intratumor injection. In addition, we find that HPV-associated (HPV+) HNSCCs are associated with increased TAS2R14 expression. Lidocaine treatment may benefit these patients, warranting future clinical studies.
Collapse
Affiliation(s)
- Zoey A Miller
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Pharmacology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Arielle Mueller
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - TaeBeom Kim
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer F Jolivert
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ray Z Ma
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sahil Muthuswami
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - April Park
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Derek B McMahon
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin T Nead
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ryan M Carey
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Robert J Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Yang Y, Yu L, Zhu T, Xu S, He J, Mao N, Liu Z, Wang D. Neuroprotective effects of Lycium barbarum polysaccharide on light-induced oxidative stress and mitochondrial damage via the Nrf2/HO-1 pathway in mouse hippocampal neurons. Int J Biol Macromol 2023; 251:126315. [PMID: 37582438 DOI: 10.1016/j.ijbiomac.2023.126315] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/17/2023]
Abstract
Light at night (LAN) induced cognitive impairment associated with oxidative stress in mice has been reported. Lycium barbarum polysaccharide (LBP) exhibits anti-tumor, anti-oxidant and neuroprotective effects, yet the neuroprotective effect on light-induced neuron damage still unclear. Here, mice exposed to LAN displayed cognitive impairment and depressive like behavior, which was reversed by LBP treatment. Meanwhile, LBP alleviated light-induced higher apoptosis and mitochondrial damage in HT-22 cells. Also, LBP prevented the decreased of mitochondrial membrane permeabilization (MMP) level in light-treated cells. Additionally, LBP demonstrated its antioxidant potential by reducing ROS production and malondialdehyde (MDA) level, while simultaneously enhancing the levels of superoxide dismutase (SOD) and glutathione peroxidases (GSH-Px) in both light-treated mice and HT-22 cells. Furthermore, the mRNA and protein expression of Nrf2 (NF-E2-related factor 2), heme oxygenease-1 (HO-1), and NAD(P)H quinone oxidoreductase (NQO1) were decreased in both light-treated mice and cells. Additionally, LBP treatment reversed light-induced the inhibition of Nrf2/HO-1 signaling pathway in both mice and cells. Moreover, Nrf2 antagonist ML385 significantly eliminated the neuroprotection of LBP on cell apoptosis, oxidative stress and mitochondrial damage in light-treated cells. These results indicate that LBP can rescue light-induced neurotoxicity in mice and HT-22 cells by activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Yang Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Lin Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tianyu Zhu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Shuwen Xu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jin He
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ningning Mao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health & Food Safety, Institute of Immunology, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
10
|
Xie M, Wu H, Bian J, Huang S, Xia Y, Qin Y, Yan Z. Synthesis and biological evaluation of capsaicin analogues as antioxidant and neuroprotective agents. RSC Adv 2023; 13:32150-32159. [PMID: 37920757 PMCID: PMC10619477 DOI: 10.1039/d3ra05107b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023] Open
Abstract
Capsaicin and its analogues 3a-3q were designed and synthesized as potential new antioxidant and neuroprotective agents. Many analogues exhibited good antioxidant effects, and some showed more potent free radical scavenging activities than the positive drug quercetin (IC50 = 8.70 ± 1.75 μM for DPPH assay and 13.85 ± 2.87 μM for ABTS assay, respectively). The phenolic hydroxyl of capsaicin analogues was critical in determining antioxidant activity. Among these compounds, 3k displayed the most potent antioxidant activity. Cell vitality tests revealed that the representative compound 3k was good at protecting cells from H2O2-induced oxidative damage at low concentrations (cell viability increased to 90.0 ± 5.5% at 10 μM). In addition, the study demonstrated that 3k could reduce intracellular ROS accumulation and increase GSH levels to prevent H2O2-induced oxidative stress in SY5Y cells. In the mitochondrial membrane potential assay, 3k significantly increased the MMP level of SY5Y cells treated with H2O2 and played an anti-neuronal cell death role. These results provide a promising strategy to develop novel capsaicin analogues as potential antioxidant and neuroprotective agents.
Collapse
Affiliation(s)
- Mao Xie
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Huixian Wu
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
- Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| | - Jing Bian
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing Jiangsu China
| | - Shutong Huang
- Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| | - Yuanzheng Xia
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing Jiangsu China
| | - Yujun Qin
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing Jiangsu China
| | - Zhiming Yan
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
- Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| |
Collapse
|
11
|
Zhang C, Liu J, Wang X, Li E, Song M, Yang Y, Qin C, Qin J, Chen L. Comprehensive transcriptional and metabolomic analysis reveals the neuroprotective mechanism of dietary gamma-aminobutyric acid response to hypoxic stress in the Chinese mitten crab (Eriocheir sinensis). FISH & SHELLFISH IMMUNOLOGY 2023; 135:108663. [PMID: 36898515 DOI: 10.1016/j.fsi.2023.108663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/07/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
Hypoxia is one of the serious stress challenges that aquatic animals face throughout their life. Our previous study found that hypoxia stress could induce neural excitotoxicity and neuronal apoptosis in Eriocheir sinensis, and observed that gamma-aminobutyric acid (GABA) has a positive neuroprotective effect on juvenile crabs under hypoxia. To reveal the neuroprotective pathway and metabolic regulatory mechanism of GABA in E. sinensis exposed to hypoxia stress, an 8-week feeding trial and acute hypoxia challenge were performed. Subsequently, we performed a comprehensive transcriptomic and metabolomic analysis of the thoracic ganglia of juvenile crabs. Differential genes and differential metabolites were co-annotated to 11 KEGG pathways, and further significant analysis showed that only the sphingolipid signaling pathway and the arachidonic acid metabolism pathway were significantly enriched. In the sphingolipid signaling pathway, GABA treatment significantly increased long-chain ceramide content in thoracic ganglia, which exerted neuroprotective effects by activating downstream signals to inhibit hypoxia-induced apoptosis. Moreover, in the arachidonic acid metabolism pathway, GABA could increase the content of neuroprotective active substances and reduce the content of harmful metabolites by regulating the metabolism of arachidonic acid for inflammatory regulation and neuroprotection. Furthermore, the decrease of glucose and lactate levels in the hemolymph suggests the positive role of GABA in metabolic regulation. This study reveals the neuroprotective pathways and possible mechanisms of GABA in juvenile E. sinensis exposed to hypoxia stress and inspires the discovery of new targets for improving hypoxia tolerance in aquatic animals.
Collapse
Affiliation(s)
- Cong Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Jiadai Liu
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Xiaodan Wang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Erchao Li
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Mingqi Song
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Yiwen Yang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Chuanjie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Sichuan, 641100, PR China
| | - Jianguang Qin
- College of Science and Engineering, Flinders University, Adelaide, SA, 5001, Australia
| | - Liqiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China.
| |
Collapse
|
12
|
Jiang C, Hu Y, Hou X, Qiu J. Neuroprotective effect of a novel brain-derived peptide, HIBDAP, against oxygen-glucose deprivation through inhibition of apoptosis in PC12 cells. Mol Biol Rep 2023; 50:3045-3051. [PMID: 36680622 DOI: 10.1007/s11033-023-08248-0] [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: 10/22/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023]
Abstract
BACKGROUND The effect of a novel brain-derived peptide, hypoxic-ischemic brain damage associated peptide (HIBDAP), on apoptosis after oxygen-glucose deprivation (OGD) in PC12 cells was investigated. METHODS The HIBDAP sequence (HSQFIGYPITLFVEKER) was coupled with the carrier peptide of the transactivator of transcription (TAT) sequence (YGRKKRRQRRR). FITC-labelled TAT-HIBDAP was observed by fluorescence microscopy. After TAT-HIBDAP treatment and OGD treatment, the PC12 cell apoptosis rate was analysed using lactate dehydrogenase (LDH) leakage and Annexin V-fluorescein isothiocyanate (FITC) assays. Mitochondrial membrane potential (ΔΨm) was examined by fluorescence microscopy. Protein expression of apoptotic factors was examined by Western blotting. RESULTS FITC-labelled TAT-HIBDAP entered the PC12 cell nucleus. Compared with the OGD group, TAT-HIBDAP at low concentrations (1 µM, 5 µM, 10 µM) significantly reduced the apoptosis rate of PC12 cells (except at 20 µM); 5 µM TAT-HIBDAP had the most obvious effect. There were remarkable increases in ΔΨm at different concentrations (1 µM, 5 µM, 10 µM, 20 µM) of TAT-HIBDAP pretreatment, and 5 µM TAT-HIBDAP also had the most obvious effect. TAT-HIBDAP reversed the increased ratio of Bax/Bcl-2 and activation of Caspase-3 induced by OGD. CONCLUSION TAT-HIBDAP is resistant to OGD-induced PC12 cell apoptosis by regulating the Bax/Bcl-2/Caspase-3 pathway, which may provide a novel therapeutic strategy for neonatal HIBD.
Collapse
Affiliation(s)
| | - Yina Hu
- Nanjing Medical University, Nanjing, China
| | - Xuewen Hou
- Dapartment of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| | - Jie Qiu
- Department of Neonatology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Lin H, Zhang Y, Dong S, Cai X, Jiang H, Fan Y, Ying K, Du B, Yu P, Yang W. Targeted Therapy of Ischemic Stroke via Crossing the Blood-Brain Barrier Using Edaravone-Loaded Multiresponsive Microgels. ACS APPLIED BIO MATERIALS 2022; 5:4165-4178. [PMID: 36083038 DOI: 10.1021/acsabm.2c00325] [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: 11/29/2022]
Abstract
Ischemic stroke, as a prevalent neurological disorder, often results in rapid increases in the production of reactive oxygen species (ROS) and inflammatory factors in the focal ischemic area. Though edaravone is an approved treatment, its effect is limited due to its weak ability to cross the blood-brain barrier (BBB) and short half-life. Other effective pharmacological treatment options are clearly lacking. In this study, PNIVDBrF-3-Eda (also named MG-3-Eda) was prepared using a thermo- and pH dual-responsive PNIVDBrF microgel. These were designed with a positively charged network, as synthesized by simultaneous quaternization cross-linking and surfactant-free emulsion copolymerization, to be loaded with the negatively charged edaravone. We then investigated whether such a targeted delivery of edaravone could provide enhanced neuroprotection. Cytotoxicity assays confirmed that the microgel (<1 mg/mL) exhibited promising cytocompatibility with no remarkable effect on cell viability, cell cycle regulation, or apoptosis levels. In vitro and in vivo experiments demonstrated that the microgels could successfully penetrate the blood-brain barrier where efficient BBB crossing was observed after disruption of the BBB due to ischemic injury. This enabled MG-3-Eda to target the cerebral ischemic area and achieve local release of edaravone. Treatment with MG-3-Eda significantly reduced the cerebral infarct area in transient middle cerebral artery occlusion (tMCAO) mice and significantly improved behavioral scores. MG-3-Eda treatment also prevented the reduction in NF200 expression, a neuronal marker protein, and attenuated microglia activation (as detected by Iba1) in the local ischemic area via local antioxidant and anti-inflammatory effects. A superior neuroprotective effect was noted for MG-3-Eda compared to that for free edaravone. Our results indicate that MG-3-Eda administration represents a clear potential treatment for cerebral ischemia via its targeted delivery of edaravone to ischemic areas where it provides significant local antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Hongwei Lin
- Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yi Zhang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Shunni Dong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou 310012, China
| | - Xiaobo Cai
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hui Jiang
- Department of Toxicology and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yang Fan
- Department of Toxicology and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Kaiyue Ying
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Binyang Du
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou 310012, China
| | - Peilin Yu
- Department of Toxicology and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| |
Collapse
|
14
|
Lao Y, Huang P, Chen J, Wang Y, Su R, Shao W, Hu W, Zhang J. Discovery of 1,2,4-triazole derivatives as novel neuroprotectants against cerebral ischemic injury by activating antioxidant response element. Bioorg Chem 2022; 128:106096. [PMID: 35985158 DOI: 10.1016/j.bioorg.2022.106096] [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: 04/29/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/02/2022]
Abstract
Acute ischemic stroke is an important cause of death and long-term disability worldwide. In this work, we have synthesized a series of derivatives with 3,5‑diaryl substituent triazole scaffolds. The derivatives showed favorable protective effective in SNP-induced oxidative stress model, of which compound 5 was the most active. In vivo experiments showed that compound 5 could ameliorate neurological deficits, attenuate infarction sizes, reduce malonaldehyde (MDA) level and increase superoxide dismutase (SOD) level in middle cerebral artery occlusion (MCAO) rats. Preliminary safety evaluation showed that compound 5 exhibited low acute toxicity in BALB/c mice (LD50 greater than 1000 mg/kg). Further investigation indicated that compound 5 was able to scavenge ROS, restore mitochondrial membrane potential and protect PC12 cells from SNP-induced apoptosis. Moreover, compound 5 could initiate transcription of antioxidant response element (ARE) and induced expressions of antioxidative enzymes. Collectively, compound 5 might have the potency of treating acute ischemic stroke.
Collapse
Affiliation(s)
- Yaoqiang Lao
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Ping Huang
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Jianwen Chen
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yang Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Ruiqi Su
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Weiyan Shao
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Wenhao Hu
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Jingxia Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
15
|
Das J, Mahammad FS, Krishnamurthy RG. An integrated chemo-informatics and in vitro experimental approach repurposes acarbose as a post-ischemic neuro-protectant. 3 Biotech 2022; 12:71. [PMID: 35223357 PMCID: PMC8847516 DOI: 10.1007/s13205-022-03130-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 01/23/2022] [Indexed: 11/26/2022] Open
Abstract
The increasing prevalence of ischemic stroke combined with limited therapeutic options highlights the compelling need for continued research into the development of future neuro-therapeutics. Death-Associated Protein Kinase 1 (DAPK1) and p53 protein-protein interaction serve as a signaling point for the convergence of apoptosis and necrosis in cerebral ischemia. In this study, we used an integrated chemo-informatics and in vitro experimental drug repurposing strategy to screen potential small-molecule inhibitors of DAPK1-p53 interaction from the United States of America Food and Drug Administration (FDA) approved drug database exhibiting post-ischemic neuroprotective and neuro-regenerative efficacy and mechanisms. The computational docking and molecular dynamics simulation of FDA-approved drugs followed by an in vitro experimental validation identified acarbose, an anti-diabetic medication and caloric restriction mimetic as a potential inhibitor of DAPK1-p53 interaction. The evaluation of post-ischemic neuroprotective and regenerative efficacy and mechanisms of action for acarbose was carried out using a set of experimental methods, including cell viability, proliferation and differentiation assays, fluorescence staining, and gene expression analysis. Post-ischemic administration of acarbose conferred significant neuroprotection against ischemia-reperfusion injury in vitro. The reduced fluorescence emission in cells stained with pS20 supported the potential of acarbose in inhibiting the DAPK1-p53 interaction. Acarbose prevented mitochondrial and lysosomal dysfunction, and favorably modulated gene expression related to cell survival, inflammation, and regeneration. BrdU staining and neurite outgrowth assay showed a significant increase in cell proliferation and differentiation in acarbose-treated group. This is the first study known to provide mechanistic insight into the post-ischemic neuroprotective and neuro-regenerative potential of acarbose. Our results provide a strong basis for preclinical studies to evaluate the safety and neuroprotective efficacy of acarbose against ischemic stroke. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13205-022-03130-5.
Collapse
Affiliation(s)
- Jyotirekha Das
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala 673601 India
| | - Fayaz Shaik Mahammad
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka 576104 India
| | | |
Collapse
|
16
|
Zhang Q, Xue H, Niu J, Tian M, Yu X. Construction of fluorescent probes for ratiometric visualization of mitochondrial depolarization based on the FRET mechanism. NEW J CHEM 2022. [DOI: 10.1039/d2nj01376b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Two fluorescent probes based on the FRET mechanism were reasonably fabricated for the visualization of mitochondrial depolarization in a ratiometric manner.
Collapse
Affiliation(s)
- Qianqian Zhang
- School of Information Science and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, China
| | - Haiyan Xue
- School of Information Science and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, China
| | - Jie Niu
- Center of Bio & Micro/Nano Functional Materials, State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, P. R. China
| | - Minggang Tian
- School of Information Science and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, China
| | - Xiaoqiang Yu
- Center of Bio & Micro/Nano Functional Materials, State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, P. R. China
| |
Collapse
|
17
|
Shao-Mei W, Li-Fang Y, Li-Hong W. Traditional Chinese medicine enhances myocardial metabolism during heart failure. Biomed Pharmacother 2021; 146:112538. [PMID: 34922111 DOI: 10.1016/j.biopha.2021.112538] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 11/02/2022] Open
Abstract
The prognosis of various cardiovascular diseases eventually leads to heart failure (HF). An energy metabolism disorder of cardiomyocytes is important in explaining the molecular basis of HF; this will aid global research regarding treatment options for HF from the perspective of myocardial metabolism. There are many drugs to improve myocardial metabolism for the treatment of HF, including angiotensin receptor blocker-neprilysin inhibitor (ARNi) and sodium glucose cotransporter 2 (SGLT-2) inhibitors. Although Western medicine has made considerable progress in HF therapy, the morbidity and mortality of the disease remain high. Therefore, HF has attracted attention from researchers worldwide. In recent years, the application of traditional Chinese medicine (TCM) in HF treatment has been gradually accepted, and many studies have investigated the mechanism whereby TCM improves myocardial metabolism; the TCMs studied include Danshen yin, Fufang Danshen dripping pill, and Shenmai injection. This enables the clinical application of TCM in the treatment of HF by improving myocardial metabolism. We systematically reviewed the efficacy of TCM for improving myocardial metabolism during HF as well as the pharmacological effects of active TCM ingredients on the cardiovascular system and the potential mechanisms underlying their ability to improve myocardial metabolism. The results indicate that TCM may serve as a complementary and alternative approach for the prevention of HF. However, further rigorously designed randomized controlled trials are warranted to assess the effect of TCM on long-term hard endpoints in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Wang Shao-Mei
- Cardiovascular Medicine Department, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, Zhejiang, China
| | - Ye Li-Fang
- Cardiovascular Medicine Department, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, Zhejiang, China
| | - Wang Li-Hong
- Cardiovascular Medicine Department, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, Zhejiang, China.
| |
Collapse
|
18
|
Kaya Tilki E, Engür Öztürk S, Özarda MG, Cantürk Z, Dikmen M. Investigation of the neuroprotective and neuritogenic effects of halotolerant Penicillium flavigenum-derived sorbicillin-like compounds on PC-12 Adh cells. Cytotechnology 2021; 73:801-813. [PMID: 34776630 DOI: 10.1007/s10616-021-00498-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is an adult-onset neurodegenerative condition caused by oxidative stress and mitochondrial malfunction. In this study, the neuroprotective and neuritogenic activity of water fraction (Sw-fr) containing sorbicillin-like active metabolites of halotolerant P. flavigenum isolated from Salt Lake in Konya, Turkey were investigated on a 6-hydroxydopamine (6-OHDA)-induced PD in vitro PC-12 Adh cell model. Firstly, Sw-fr containing sorbicillin-like active metabolites were extracted from P. flavigenum and was compared with a sorbicillin standard by liquid chromatography-mass spectrometry (LC-MS). Then, the effects of non-cytotoxic concentrations of Sw-fr on the 6-OHDA-induced PD cell model were investigated via real-time cell proliferation analysis using the RTCA DP instrument. The effects of these concentrations on mitochondrial membrane integrity, caspase-3 were investigated by flow cytometry. Neurite outgrowth analysis and immunofluorescence staining were used to explore the neuritogenic effects of neuroprotective doses. By improving PC-12 Adh cell viability, decreasing reactive oxygen species production, and reducing apoptotic cell death, 1 and 10 μg/mL Sw-fr and sorbicillin standard proved neuroprotective against 6-OHDA-induced neurotoxicity. Furthermore, 1 and 10 µg/mL Sw-fr significantly induced neurite outgrowth. As a result, sorbicillin-like active metabolites containing Sw-fr were found to have neuroprotective and neuritogenic effects. Sorbicillin-like metabolites obtained from fungi may be novel natural medicines for neurodegenerative diseases. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-021-00498-9.
Collapse
Affiliation(s)
- Elif Kaya Tilki
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, 26210 Eskisehir, Turkey
| | - Selin Engür Öztürk
- Tavas Vocational School of Health Services, Pamukkale University, Denizli, Turkey
| | - Mustafa Güçlü Özarda
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Anadolu University, 26210 Eskisehir, Turkey
| | - Zerrin Cantürk
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Anadolu University, 26210 Eskisehir, Turkey
| | - Miriş Dikmen
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, 26210 Eskisehir, Turkey
| |
Collapse
|
19
|
Umbrasas D, Arandarcikaite O, Grigaleviciute R, Stakauskas R, Borutaite V. Neuroprotective Effect of a Novel ATP-Synthase Inhibitor Bedaquiline in Cerebral Ischemia-Reperfusion Injury. Int J Mol Sci 2021; 22:9717. [PMID: 34575875 PMCID: PMC8472139 DOI: 10.3390/ijms22189717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dysfunction during ischemic stroke ultimately manifests as ATP depletion. Mitochondrial ATP synthase upon loss of mitochondrial membrane potential during ischemia rapidly hydrolyses ATP and thus contributes to ATP depletion. Increasing evidence suggests that inhibition of ATP synthase limits ATP depletion and is protective against ischemic tissue damage. Bedaquiline (BDQ) is an anti-microbial agent, approved for clinical use, that inhibits ATP synthase of Mycobacteria; however recently it has been shown to act on mitochondrial ATP synthase, inhibiting both ATP synthesis and hydrolysis in low micromolar concentrations. In this study, we investigated whether preconditioning with BDQ can alleviate ischemia/reperfusion-induced brain injury in Wistar rats after middle cerebral artery occlusion-reperfusion and whether it affects mitochondrial functions. We found that BDQ was effective in limiting necrosis and neurological dysfunction during ischemia-reperfusion. BDQ also caused inhibition of ATPase activity, mild uncoupling of respiration, and stimulated mitochondrial respiration both in healthy and ischemic mitochondria. Mitochondrial calcium retention capacity was unaffected by BDQ preconditioning. We concluded that BDQ has neuroprotective properties associated with its action on mitochondrial respiration and ATPase activity.
Collapse
Affiliation(s)
- Danielius Umbrasas
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (O.A.); (V.B.)
| | - Odeta Arandarcikaite
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (O.A.); (V.B.)
| | - Ramune Grigaleviciute
- Biological Research Center, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (R.G.); (R.S.)
| | - Rimantas Stakauskas
- Biological Research Center, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (R.G.); (R.S.)
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (O.A.); (V.B.)
| |
Collapse
|
20
|
Wang DP, Jin KY, Zhao P, Lin Q, Kang K, Hai J. Neuroprotective Effects of VEGF-A Nanofiber Membrane and FAAH Inhibitor URB597 Against Oxygen-Glucose Deprivation-Induced Ischemic Neuronal Injury. Int J Nanomedicine 2021; 16:3661-3678. [PMID: 34093011 PMCID: PMC8168836 DOI: 10.2147/ijn.s307335] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Brain ischemia is a common neurological disorder worldwide that activates a cascade of pathophysiological events involving decreases in oxygen and glucose levels. Despite substantial efforts to explore its pathogenesis, the management of ischemic neuronal injury remains an enormous challenge. Accumulating evidence suggests that VEGF modified nanofiber (NF) materials and the fatty-acid amide hydrolase (FAAH) inhibitor URB597 exert an influence on alleviating ischemic brain damage. We aimed to further investigate their effects on primary hippocampal neurons, as well as the underlying mechanisms following oxygen-glucose deprivation (OGD). METHODS Different layers of VEGF-A loaded polycaprolactone (PCL) nanofibrous membranes were first synthesized by using layer-by-layer (LBL) self-assembly of electrospinning methods. The physicochemical and biological properties of VEGF-A NF membranes, and their morphology, hydrophilicity, and controlled-release of VEGF-A were then estimated. Furthermore, the effects of VEGF-A NF and URB597 on OGD-induced mitochondrial oxidative stress, inflammatory responses, neuronal apoptosis, and endocannabinoid signaling components were assessed. RESULTS The VEGF-A NF membrane and URB597 can not only promote hippocampal neuron adhesion and viability following OGD but also exhibited antioxidant/anti-inflammatory and mitochondrial membrane potential protection. The VEGF-A NF membrane and URB597 also inhibited OGD-induced cellular apoptosis through activating CB1R signaling. These results indicate that VEGF-A could be controlled-released by LBL self-assembled NF membranes. DISCUSSION The VEGF-A NF membrane and URB597 displayed positive synergistic neuroprotective effects through the inhibition of mitochondrial oxidative stress and activation of CB1R/PI3K/AKT/BDNF signaling, suggesting that a VEGF-A loaded NF membrane and the FAAH inhibitor URB597 could be of therapeutic value in ischemic cerebrovascular diseases.
Collapse
Affiliation(s)
- Da-Peng Wang
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai, 200065, People’s Republic of China
| | - Kai-Yan Jin
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai, 200065, People’s Republic of China
| | - Peng Zhao
- Institute for Translational Medicine, Institute for Biomedical Engineering and Nanoscience, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, People’s Republic of China
| | - Qi Lin
- Department of Pharmacy, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Kai Kang
- Department of Research and Surveillance Evaluation, Shanghai Center for Health Promotion, Shanghai, 200040, People’s Republic of China
| | - Jian Hai
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai, 200065, People’s Republic of China
| |
Collapse
|
21
|
Zhang T, Li Z, Qin Z, Cao Y, Shan T, Fang Y, Tang L, Jia N, Jia J, Jin Z, Xu T, Li Y. Neuroprotection of Chikusetsu saponin V on transient focal cerebral ischemia/reperfusion and the underlying mechanism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153516. [PMID: 33639592 DOI: 10.1016/j.phymed.2021.153516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 01/20/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Oxidative stress and frequently unwanted alterations in mitochondrial structure and function are key aspects of the pathological cascade in transient focal cerebral ischemia. Chikusetsu saponin V (CHS V), a major component of saponins from Panax japonicas, can attenuate H2O2-induced oxidative stress in SH-SY5Y cells. PURPOSE The aim of the present study was to investigate the neuroprotective effects and the possible underlying mechanism of CHS V on transient focal cerebral ischemia/reperfusion. METHODS Mice with middle cerebral artery occlusion (MCAO) and cultured cortical neurons exposed to oxygen glucose deprivation (OGD) were used as in vivo and in vitro models of cerebral ischemia, respectively. The neurobehavioral scores, infarction volumes, H&E staining and some antioxidant levels in the brain were evaluated. The occurrence of neuronal death was estimated. Total and mitochondrial reactive oxygen species (ROS) levels, as well as mitochondrial potential were measured using flow cytometry analysis. Mitochondrial structure and respiratory activity were also examined. Protein levels were investigated by western blotting and immunohistochemistry. RESULTS CHS V effectively attenuated cerebral ischemia/reperfusion (CI/R) injury, including improving neurological deficits, shrinking infarct volume and reducing the number of apoptotic cells. Furthermore, CHS V treatment remarkably increased antioxidant levels and reduced ROS levels and mitochondrial damage by enhancing the expression and deacetylation of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) by activating AMPK and SIRT-1, respectively. CONCLUSION Our data demonstrated that CHS V prevented CI/R injury by suppressing oxidative stress and mitochondrial damage through the modulation of PGC-1α with AMPK and SIRT-1.
Collapse
Affiliation(s)
- Tiejun Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengjun Li
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Zhou Qin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Cao
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tikun Shan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Fang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linqiao Tang
- Core Facility, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Jia
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jing Jia
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Zhaohui Jin
- Core Facility, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ting Xu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
22
|
Transmembrane Prolyl 4-Hydroxylase is a Novel Regulator of Calcium Signaling in Astrocytes. eNeuro 2021; 8:ENEURO.0253-20.2020. [PMID: 33298456 PMCID: PMC7814479 DOI: 10.1523/eneuro.0253-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/09/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022] Open
Abstract
Prolyl 4-hydroxylases (P4Hs) have vital roles in regulating collagen synthesis and hypoxia response. A transmembrane P4H (P4H-TM) is a recently identified member of the family. Biallelic loss of function P4H-TM mutations cause a severe autosomal recessive intellectual disability syndrome in humans, but functions of P4H-TM are essentially unknown at cellular level. Our microarray data on P4h-tm -/- mouse cortexes where P4H-TM is abundantly expressed indicated expression changes in genes involved in calcium signaling and expression of several calcium sequestering ATPases was upregulated in P4h-tm -/- primary mouse astrocytes. Cytosolic and intraorganellar calcium imaging of P4h-tm -/- cells revealed that receptor-operated calcium entry (ROCE) and store-operated calcium entry (SOCE) and calcium re-uptake by mitochondria were compromised. HIF1, but not HIF2, was found to be a key mediator of the P4H-TM effect on calcium signaling. Furthermore, total internal reflection fluorescence (TIRF) imaging showed that calcium agonist-induced gliotransmission was attenuated in P4h-tm -/- astrocytes. This phenotype was accompanied by redistribution of mitochondria from distal processes to central parts of the cell body and decreased intracellular ATP content. Our data show that P4H-TM is a novel regulator of calcium dynamics and gliotransmission.
Collapse
|
23
|
AMPK-SIRT1-PGC1α Signal Pathway Influences the Cognitive Function of Aged Rats in Sevoflurane-Induced Anesthesia. J Mol Neurosci 2020; 70:2058-2067. [PMID: 32514740 DOI: 10.1007/s12031-020-01612-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022]
Abstract
To understand the effect of AMP-activated protein kinase (AMPK)-SIRT1 (silent information regulator 1)-PPARγ coactivator-1α (PGC1α) signaling pathway on the cognitive function of sevoflurane-anesthetized aged rats. Aged rats were divided into Normal group, Sevo group (Sevoflurane anesthesia), Sevo + AICAR (the AMPK activator) group, Sevo + EX527 group (the AMPK inhibitor), and Sevo + AICAR + EX527 group. The cognitive function of rats was determined by the Morris water maze. Hippocampal neuronal apoptosis was evaluated by TUNEL and Fluoro-Jade C (FJC) staining, and the expression of cleaved caspase-3 was detected by immunohistochemistry. ROS, SOD, and MDA levels and the fluorescence intensity of GFAP in the hippocampus were assayed. The mitochondrial membrane potential (MMP), mitochondrial mass, ATP level, and the expression of AMPK-SIRT1-PGC1α were determined by the corresponding methods. Rats in the Sevo group manifested significant extension in the escape latency, with fewer platform crossings; and meanwhile, the apoptotic rate, the number of FJC-positive cells, and the fluorescence intensity of GFAP of neurons were elevated, with up-regulation of cleaved caspase-3. Moreover, the level of MDA and ROS was increased evidently, with significant down-regulation of SOD activity, ATP, mitochondrial mass and MMP levels, and AMPK, SIRT1 and PGC-1α protein expressions. However, sevoflurane-induced changes above were improved after the administration of AICAR, and EX527 could reverse AICAR-induced improvements in Sevo-anesthetized aged rats. Activating AMPK-SIRT1-PGC1α pathway can improve the cognitive function and mitigate the neuronal injury in Sevo-anesthetized aged rats by antagonizing the oxidative stress and maintaining the mitochondrial function.
Collapse
|
24
|
MCL-1 Matrix maintains neuronal survival by enhancing mitochondrial integrity and bioenergetic capacity under stress conditions. Cell Death Dis 2020; 11:321. [PMID: 32371858 PMCID: PMC7200794 DOI: 10.1038/s41419-020-2498-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022]
Abstract
Mitochondria play a crucial role in neuronal survival through efficient energy metabolism. In pathological conditions, mitochondrial stress leads to neuronal death, which is regulated by the anti-apoptotic BCL-2 family of proteins. MCL-1 is an anti-apoptotic BCL-2 protein localized to mitochondria either in the outer membrane (OM) or inner membrane (Matrix), which have distinct roles in inhibiting apoptosis and promoting bioenergetics, respectively. While the anti-apoptotic role for Mcl1 is well characterized, the protective function of MCL-1 Matrix remains poorly understood. Here, we show MCL-1OM and MCL-1Matrix prevent neuronal death through distinct mechanisms. We report that MCL-1Matrix functions to preserve mitochondrial energy transduction and improves respiratory chain capacity by modulating mitochondrial oxygen consumption in response to mitochondrial stress. We show that MCL-1Matrix protects neurons from stress by enhancing respiratory function, and by inhibiting mitochondrial permeability transition pore opening. Taken together, our results provide novel insight into how MCL-1Matrix may confer neuroprotection under stress conditions involving loss of mitochondrial function.
Collapse
|
25
|
Postischemic supplementation of folic acid improves neuronal survival and regeneration in vitro. Nutr Res 2020; 75:1-14. [DOI: 10.1016/j.nutres.2019.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/05/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
|
26
|
Zhang Y, Cao M, Wu Y, Wang J, Zheng J, Liu N, Yang N, Liu Y. Improvement in mitochondrial function underlies the effects of ANNAO tablets on attenuating cerebral ischemia-reperfusion injuries. JOURNAL OF ETHNOPHARMACOLOGY 2020; 246:112212. [PMID: 31494200 DOI: 10.1016/j.jep.2019.112212] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/10/2019] [Accepted: 08/31/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE ANNAO tablets derive from Chinese classical prescriptions of Angong Niuhuang Pills with modified compositions, which have been singly or combined used for stoke associated neurological disorders. However the underlying mechanism is not yet well-defined, the present study investigated its anti-ischemic effects in rat middle cerebral artery occlusion (MCAO) model and focused on mitochondrial quality control. MATERIALS AND METHODS Rats were subjected to 2 h of brain ischemia followed by 1 day or up to 7 days of reperfusion. Vehicle, ANNAO tablets or Edaravone were given at 1h after the start of reperfusion for 1 day or successive 7 days in MCAO rats. For the behavior assessment, Longa test and modified Neurological Severity Scores (m NSS) test were performed. Following the behavioral assessment, we assessed the protein expressions related to mitochondrial function. Moreover, we also assessed the neuroprotective effects of ANNAO tablets by immunohistochemical analysis. RESULTS Compared with sham rats, ANNAO tablets improved the behavioral performance and decreased the infarction volume in the MCAO rats. Western blotting results showed that ANNAO tablets altered the expression level of multiple proteins related to mitochondrial function, elevated the ratio of Bcl-2/Bax and inhibited the apoptosis. Additionally, ANNAO tablets increased the number of NeuN positive neurons. CONCLUSIONS The obtained data demonstrated that ANNAO tablets exhibited an obvious anti-cerebral ischemia-reperfusion effect, which could be attributed to the improvement of mitochondrial quality control.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Mingyue Cao
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Youming Wu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Jun Wang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Ji Zheng
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Nasi Liu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Nan Yang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| | - Yanyong Liu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
27
|
Jiao J, Wang Y, Ren P, Sun S, Wu M. Necrosulfonamide Ameliorates Neurological Impairment in Spinal Cord Injury by Improving Antioxidative Capacity. Front Pharmacol 2020; 10:1538. [PMID: 31998134 PMCID: PMC6962303 DOI: 10.3389/fphar.2019.01538] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/27/2019] [Indexed: 12/17/2022] Open
Abstract
Currently, there is no efficient therapy for spinal cord injury (SCI). Anoxemia after SCI is a key problem, which leads to tissue destruction, while hypoxia after SCI induces cell injury along with inflammation. Mixed-lineage kinase domain-like protein (MLKL) is a critical signal molecule of necroptosis, and mitochondrial dysfunction is regarded as one of the most pivotal events after SCI. Based on the important role of MLKL in cell damage and potential role of mitochondrial dysfunction, our study focuses on the regulation of MLKL by Necrosulfonamide (NSA) in mitochondrial dysfunction of oxygen-glucose deprivation (OGD)-induced cell damage and SCI-mice, which specifically blocks the MLKL. Our results showed that NSA protected against a decrease in the mitochondrial membrane potential, adenosine triphosphate, glutathione, and superoxide dismutase levels and an increase in reactive oxygen species and malonyldialdehyde levels. NSA also improved the locomotor function in SCI-mice and OGD-induced spinal neuron injury through inhibition of MLKL activation independently of receptor-interacting protein kinase 3 (RIP3) phosphorylation. Besides the protective effects, NSA exhibited a therapeutic window. The optimal treatment time was within 12 h after the injury in the SCI-mice model. In conclusion, our data suggest a close association between the NSA level inhibiting p-MLKL independently of RIP3 phosphorylation and induction of neurological impairment by improving antioxidative capacity after SCI. NSA ameliorates neurological impairment in SCI through inhibiting MLKL-dependent necroptosis. It also provides a theoretical basis for further research and application of NSA in the treatment of SCI.
Collapse
Affiliation(s)
- Jianhang Jiao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Yang Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Pengfei Ren
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Shicai Sun
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Minfei Wu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
28
|
Cai M, Yang Q, Li G, Sun S, Chen Y, Tian L, Dong H. Activation of cannabinoid receptor 1 is involved in protection against mitochondrial dysfunction and cerebral ischaemic tolerance induced by isoflurane preconditioning. Br J Anaesth 2019; 119:1213-1223. [PMID: 29045576 DOI: 10.1093/bja/aex267] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2017] [Indexed: 12/13/2022] Open
Abstract
Background Isoflurane preconditioning (IPC) induces cerebral ischaemic tolerance, but the mechanism remains poorly understood. The aim of this study was to determine changes in mitochondrial function in the brain after IPC, and whether the cannabinoid receptor 1 (CB1R) could be involved in the mechanism of mitochondrial protection mediated by IPC. Methods Adult male Sprague-Dawley rats were pretreated with isoflurane 2% for 1 h day -1 , for 5 days consecutively, and then subjected to 120 min right middle cerebral artery occlusion. Cannabinoid receptor 1 expression in the cellular and mitochondrial membrane was measured. The CB1R agonist HU-210 was administered alone, or the antagonists AM251 and SR141716A were given to the animals before each preconditioning. Neurological scores, infarct volume, apoptosis, and mitochondrial function were examined after middle cerebral artery occlusion. Results Expression of CB1R on cellular and mitochondrial membranes was increased 6 h after preconditioning. Both IPC and HU-210 administration before middle cerebral artery occlusion improved neurological outcomes and reduced infarct volume. Isoflurane preconditioning increased the expression of the anti-apoptotic proteins Bcl-2 and Bcl-X L and reduced apoptosis in neurones. Isoflurane preconditioning and HU-210 also markedly preserved the activity of respiratory chain complexes, reduced mitochondrial radical generation, preserved mitochondrial membrane potential, and inhibited mitochondrial permeability transition pore opening. Cannabinoid receptor 1 antagonists abolished the improvement in mitochondrial function and the neuroprotective effects induced by IPC. Conclusions Our results indicate that IPC elicits brain ischaemic tolerance and mitochondrial protection by activating the CB1R, which provides a new mechanism for IPC-induced neuroprotection against cerebral ischaemia.
Collapse
Affiliation(s)
- M Cai
- Department of Anaesthesiology and Perioperative Medicine.,Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Q Yang
- Department of Anaesthesiology and Perioperative Medicine
| | - G Li
- Department of Anaesthesiology and Perioperative Medicine
| | - S Sun
- Department of Anaesthesiology and Perioperative Medicine
| | - Y Chen
- Department of Anaesthesiology and Perioperative Medicine
| | - L Tian
- Department of Anaesthesiology and Perioperative Medicine
| | - H Dong
- Department of Anaesthesiology and Perioperative Medicine
| |
Collapse
|
29
|
Gu N, Wang J, Di Z, Liu Z, Jia X, Yan Y, Chen X, Zhang Q, Qian Y. The Effects of Intelectin-1 on Antioxidant and Angiogenesis in HUVECs Exposed to Oxygen Glucose Deprivation. Front Neurol 2019; 10:383. [PMID: 31040819 PMCID: PMC6477047 DOI: 10.3389/fneur.2019.00383] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/28/2019] [Indexed: 12/18/2022] Open
Abstract
Objective: Ischemic stroke leads to cellular death and tissue damage by depriving the areas of glucose and oxygen supplies. The effective treatment of stroke remains a challenge for modern medicine. This study used an oxygen-glucose deprivation (OGD) model of human umbilical vein endothelial cells (HUVECs) to mimic ischemic injuries and explored the role and mechanism of intelectin-1. Methods: Intelectin-1 was transduced into the HUVECs using a lentiviral vector. The PI3K/Akt signaling was examined in intelectin-induced eNOS phosphorylation. The PI3K inhibitor LY294002 was dealed in HUVECs. Results: Our results demonstrated an increase in capillary density, decrease in apoptotic cells, and increase in HIF-1α protein expression following intelectin-1 treatment. Real-time PCR and Western blotting revealed the increased intelectin-1 expression alongside eNOS and Akt phosphorylation with enhanced bcl-2 expression under OGD. Capillary density decreased significantly after LY294002 treatment. Conclusion: These results suggest intelectin-1 promotes angiogenesis, inhibits oxidative stress and reduces apoptosis by stimulating the Akt-eNOS signaling pathway in response to ischemia in vitro.
Collapse
Affiliation(s)
- Naibing Gu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Jun Wang
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Zhengli Di
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Zhiqin Liu
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Xiaotao Jia
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Yu'e Yan
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Xiaoshan Chen
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Quanzeng Zhang
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Yihua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
30
|
Song XY, Hu JF, Wu DH, Ji HJ, Chen NH. IMM-H004, a Novel Coumarin Derivative Compound, Inhibits H 2O 2-Induced Neurotoxicity via Antioxidant and Antiapoptosis in PC12 Cells. J Stroke Cerebrovasc Dis 2018; 27:3396-3403. [PMID: 30243729 DOI: 10.1016/j.jstrokecerebrovasdis.2018.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/06/2018] [Accepted: 05/19/2018] [Indexed: 11/17/2022] Open
Abstract
IMM-H004 [7-hydroxy-5-methoxy-4-methyl-3-(4-methylpiperazin-1-yl)-coumarin] is a novel derivative of coumarin, which played neuroprotective roles in brain ischemia in rats in previous studies. Although antiapoptosis and improving synapsis structure were proved, the effects and mechanisms of IMM-H004 in brain ischemia need further study. In this paper, the effect of IMM-H004 on H2O2-induced neurotoxicity in pheochromocytoma (PC12) cells was researched. Morphological observation, MTT method and PI/Hoechst staining were used to indicate cell viability and apoptosis. JC-1 and DCFH-DA were used to test mitochondrial membrane potential (MMP) and reactive oxygen species (ROS), respectively. The antioxidative activity was detected by Glutathione (GSH) and Total Antioxidant Capacity (TAC) Assay kits. Western blot was used to test apoptosis related proteins. Our results showed that treatment with 1-10 μM IMM-H004 markedly increased cell viability and decreased cell apoptosis induced by H2O2. Moreover, 1-10 μM IMM-H004 could enhance MMP and protect mitochondrial function. 1-10 μM IMM-H004 also could lower the ROS and raise the GSH and TAC level. Furthermore, 1-10 μM IMM-H004 could decrease the ratio of Bax/Bcl-2 and increase the ratio of p-AKT/AKT, which were related to apoptosis and survival. All these indicated that IMM-H004 protects PC12 cells against H2O2-induced neurotoxicity. Antioxidative and antiapoptosis may be the mechanisms of IMM-H004 in brain ischemia. These studies indicate that IMM-H004 might be a potential drug for treatment brain ischemia.
Collapse
Affiliation(s)
- Xiu-Yun Song
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jin-Feng Hu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong-Hui Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hai-Jie Ji
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
31
|
Inhibition of PTEN protects PC12 cells against oxygen-glucose deprivation induced cell death through mitoprotection. Brain Res 2018; 1692:100-109. [DOI: 10.1016/j.brainres.2018.05.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/24/2018] [Accepted: 05/18/2018] [Indexed: 01/06/2023]
|
32
|
Li B, Chen X, Yang W, He J, He K, Xia Z, Zhang J, Xiang G. Single-walled carbon nanohorn aggregates promotes mitochondrial dysfunction-induced apoptosis in hepatoblastoma cells by targeting SIRT3. Int J Oncol 2018; 53:1129-1137. [PMID: 29956732 PMCID: PMC6065448 DOI: 10.3892/ijo.2018.4459] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/11/2018] [Indexed: 01/10/2023] Open
Abstract
Single-walled carbon nanohorns (SWNHs) can accumulate in a variety of cell types or tissues and exert biological effects, which have been demonstrated to induce apoptosis in hepatoblastoma cells. However, the role and molecular mechanisms of SWNHs remain unclear. The mitochondrion is an important subcellular structure and may contribute to apoptosis that is induced by SWNHs in hepatoblastoma cells. To address this question, the mitochondrial function of HepG2 or L02 cells that were treated with SWNHs was examined. The results indicated that SWNHs were able to decrease the mitochondrial membrane potential and suppress the activity of the Na+/K+-ATPase. Secondly, HepG2 cells and L02 cells were treated with SWNHs in vivo and in vitro. The expression of mitochondrial-associated proteins [acyl-CoA synthetase short chain family member 1, Bax, cytochrome C (CYT-C), sodium channel epithelial 1α subunit, sirtuin 3 (SIRT3) and voltage-dependent anion channel 1] was analyzed by western blotting and immunohistochemical staining. The results revealed that SWNH treatment was able to alter the expression of multiple mitochondrial apoptotic pathway-associated proteins in HepG2 cells. SWNH treatment was able upregulate the expression of SIRT3, CYT-C and VDAC1 and downregulate the expression of AceCS2, but it had a more stable effect on SIRT3. However, similar findings were not observed in L02 cells. Therefore, the data from the present study indicated that SWNHs might be used as a safe anticancer agent, where it is able to trigger mitochondrial dysfunction-induced apoptosis by upregulating SIRT3 expression in HepG2 cells.
Collapse
Affiliation(s)
- Bowei Li
- Department of Postgraduate Studies, The Second Clinical College of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaoxun Chen
- Department of Gastrointestinal Surgery, The Guigang City People's Hospital, Guigang, Guangxi 537100, P.R. China
| | - Wenbin Yang
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jingliang He
- Department of General Surgery, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, Guangdong 528300, P.R. China
| | - Ke He
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Zhenglin Xia
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Jinqian Zhang
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Guoan Xiang
- Department of Postgraduate Studies, The Second Clinical College of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
33
|
Baek JH, Lee J, Yun HS, Lee CW, Song JY, Um HD, Park JK, Park IC, Kim JS, Kim EH, Hwang SG. Kinesin light chain-4 depletion induces apoptosis of radioresistant cancer cells by mitochondrial dysfunction via calcium ion influx. Cell Death Dis 2018; 9:496. [PMID: 29717133 PMCID: PMC5931584 DOI: 10.1038/s41419-018-0549-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 01/18/2023]
Abstract
Kinesins act as molecular microtubule-dependent motor proteins and have various important cellular functions related to cell division, intracellular transport, and membrane trafficking. However, the function of kinesin light chain 4 (KLC4) in cancer, especially radioresistance, has not been previously described. Thus, we investigated KLC4 function in lung cancer cells and radioresistant R-H460 cells by analyzing alterations in radiosensitivity after gene knockdown with siRNA and by evaluating cellular phenotypes and xenograft tumor growth. KLC4 was upregulated in human lung cancer cell lines. Moreover, in paired clinical specimens of lung cancer patients, KLC4 expression was significantly higher in tumor tissues than in paired adjacent normal tissues. Fluorescence-activated cell sorting (FACS) analysis showed that apoptosis rates and cleaved poly (ADP-ribose) polymerase (PARP) and cleaved caspase-3 levels in KLC4-knockdown lung cancer cells were significantly increased compared with those in control cells. Colony formation decreased as the radiation dose increased in KLC4-knockdown lung cancer cells, demonstrating an essential role for KLC4 in radioresistance. Importantly, KLC4 silencing suppressed tumor growth in an in vivo xenograft model, accompanied by increased apoptosis. Finally, KLC4-knockdown cells exhibited impaired mitochondrial respiration, increased mitochondrial reactive oxygen species production, and enhanced mitochondrial calcium uptake, resulting in mitochondrial dysfunction. Thus, KLC4 as a kinesin superfamily-targeted therapy may represent a novel, effective anticancer strategy, particularly for patients showing radioresistance.
Collapse
Affiliation(s)
- Jeong-Hwa Baek
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 440-746, Korea
| | - Janet Lee
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Hong Shik Yun
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 440-746, Korea
| | - Jie-Young Song
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Hong-Duck Um
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Jong Kuk Park
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - In-Chul Park
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Jae-Sung Kim
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Eun Ho Kim
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea.
| | - Sang-Gu Hwang
- Division of Applied Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea.
| |
Collapse
|
34
|
Shapla UM, Solayman M, Alam N, Khalil MI, Gan SH. 5-Hydroxymethylfurfural (HMF) levels in honey and other food products: effects on bees and human health. Chem Cent J 2018; 12:35. [PMID: 29619623 PMCID: PMC5884753 DOI: 10.1186/s13065-018-0408-3] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/27/2018] [Indexed: 02/07/2023] Open
Abstract
An organic compound known as 5-hydroxymethylfurfural (HMF) is formed from reducing sugars in honey and various processed foods in acidic environments when they are heated through the Maillard reaction. In addition to processing, storage conditions affect the formation HMF, and HMF has become a suitable indicator of honey quality. HMF is easily absorbed from food through the gastrointestinal tract and, upon being metabolized into different derivatives, is excreted via urine. In addition to exerting detrimental effects (mutagenic, genotoxic, organotoxic and enzyme inhibitory), HMF, which is converted to a non-excretable, genotoxic compound called 5-sulfoxymethylfurfural, is beneficial to human health by providing antioxidative, anti-allergic, anti-inflammatory, anti-hypoxic, anti-sickling, and anti-hyperuricemic effects. Therefore, HMF is a neo-forming contaminant that draws great attention from scientists. This review compiles updated information regarding HMF formation, detection procedures, mitigation strategies and effects of HMF on honey bees and human health.
Collapse
Affiliation(s)
- Ummay Mahfuza Shapla
- Laboratory of Preventive and Integrative Bio-medicine, Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Md Solayman
- Laboratory of Preventive and Integrative Bio-medicine, Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh. .,Department of Biochemistry, Primeasia University, Banani, 1213, Bangladesh.
| | - Nadia Alam
- School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Md Ibrahim Khalil
- Laboratory of Preventive and Integrative Bio-medicine, Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh.,School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
35
|
Shukla V, Fuchs P, Liu A, Cohan CH, Dong C, Wright CB, Perez-Pinzon MA, Dave KR. Recurrent Hypoglycemia Exacerbates Cerebral Ischemic Damage in Diabetic Rats via Enhanced Post-Ischemic Mitochondrial Dysfunction. Transl Stroke Res 2018; 10:78-90. [PMID: 29569040 DOI: 10.1007/s12975-018-0622-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/06/2018] [Accepted: 03/08/2018] [Indexed: 12/17/2022]
Abstract
Diabetes significantly increases the risk of stroke and post-stroke mortality. Recurrent hypoglycemia (RH) is common among diabetes patients owing to glucose-lowering therapies. Earlier, we showed that RH in a rat model of insulin-dependent diabetes exacerbates cerebral ischemic damage. Impaired mitochondrial function has been implicated as a central player in the development of cerebral ischemic damage. Hypoglycemia is also known to affect mitochondrial functioning. The present study tested the hypothesis that prior exposure of insulin-treated diabetic (ITD) rats to RH exacerbates brain damage via enhanced post-ischemic mitochondrial dysfunction. In a rat model of streptozotocin-induced diabetes, we evaluated post-ischemic mitochondrial function in RH-exposed ITD rats. Rats were exposed to five episodes of moderate hypoglycemia prior to the induction of cerebral ischemia. We also evaluated the impact of RH, both alone and in combination with cerebral ischemia, on cognitive function using the Barnes circular platform maze test. We observed that RH exposure to ITD rats leads to increased cerebral ischemic damage and decreased mitochondrial complex I activity. Exposure of ITD rats to RH impaired spatial learning and memory. Our results demonstrate that RH exposure to ITD rats potentially increases post-ischemic damage via enhanced post-ischemic mitochondrial dysfunction.
Collapse
Affiliation(s)
- Vibha Shukla
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Perry Fuchs
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Allen Liu
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Charles H Cohan
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Chuanhui Dong
- Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Clinton B Wright
- Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA. .,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA. .,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA. .,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
36
|
Protective Effects of Spatholobi Caulis Extract on Neuronal Damage and Focal Ischemic Stroke/Reperfusion Injury. Mol Neurobiol 2017; 55:4650-4666. [DOI: 10.1007/s12035-017-0652-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/11/2017] [Indexed: 01/26/2023]
|
37
|
Wu J, Hu W, Gong Y, Wang P, Tong L, Chen X, Chen Z, Xu X, Yao W, Zhang W, Huang C. Current pharmacological developments in 2,3,4',5-tetrahydroxystilbene 2-O-β-D-glucoside (TSG). Eur J Pharmacol 2017; 811:21-29. [PMID: 28545778 DOI: 10.1016/j.ejphar.2017.05.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 05/11/2017] [Accepted: 05/17/2017] [Indexed: 12/18/2022]
Abstract
2,3,4',5-tetrahydroxystilbene 2-O-β-D-glucoside (TSG), a resveratrol analog with glucoside, is purified from a traditional Chinese herbal medicine polygonum multiflorum. It has been extensively studied in last decade and known to exert strong anti-inflammatory, anti-oxidative, anti-apoptotic, and free radical scavenging activities, and therefore has been listed as a potential agent for disease therapies. Recent studies extend well-beyond effects of TSG on the injury of neurons, cardiomyocytes and endothelial cells, and report important functions of TSG in a lot of pathophysiological conditions. For example, TSG has been shown to prevent the production of pro-inflammatory cytokines in microglia and macrophages in vitro, and ameliorate pro-inflammatory responses in animal models with neurodegeneration, atherosclerosis, and rat paw or ear oedema. TSG can prevent the proliferation of vascular smooth cells, gastrointestinal dysfunctions, platelet aggregation, osteoblastic injury, diabetic nephropathy and melanogenesis. TSG is also indicated to facilitate long-term potentiation and learning and memory in both normal and pathological conditions. These effects to some extent enrich the understanding about the role of TSG in disease prevention and therapy. However, to date, we still have no outlined knowledges about the pharmacological effects of TSG, though the role of TSG in aging and Alzheimer's disease has been reviewed in recent years. Here, we summarize the current pharmacological developments of TSG as well as its possible mechanisms in disease prevention and therapy, aiming to push the understanding about the protective role of TSG as well as its preclinical assessment of novel applications.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Wenfeng Hu
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Yu Gong
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Peng Wang
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Lijuan Tong
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Xiangfan Chen
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, the Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu, China
| | - Xiaole Xu
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Wenjuan Yao
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Wei Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
38
|
Improvement of mitochondrial function mediated the neuroprotective effect of 5-(4-hydroxy-3-dimethoxybenzylidene)-2-thioxo-4-thiazolidinone in rats with cerebral ischemia-reperfusion injuries. Oncotarget 2017; 8:61193-61202. [PMID: 28977856 PMCID: PMC5617416 DOI: 10.18632/oncotarget.18048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/25/2017] [Indexed: 11/25/2022] Open
Abstract
Deficits in mitochondrial function is a critical inducement in the major pathways that drive neuronal cell death in ischemic process particularly. Drugs target to improve the mitochondrial function may be a feasible therapeutic choice in treatment with ischemic diseases. In the present study, we investigated whether 5-(4-hydroxy-3-dimethoxybenzylidene)-2-thioxo-4-thiazolidinone (RD-1), a compound derived from rhodanine, could protect against ischemic neuronal damage via improving mitochondrial function. We tested the neuroprotective effect of RD-1 both in rats modeled by middle cerebral artery occlusion reperfusion in vivo and in primary cortical neurons subjected to hypoxia/reperfusion injury in vitro. Results showed that treatment with RD-1 for 14 days remarkably reduced infarct size, decreased neurological deficit score and accelerated the recovery of somatosensory function in vivo. Meanwhile, RD-1 also increased the cellular viability after 48 h treatment in vitro. In addition, RD-1 protected the primary cortical neurons against mitochondrial damage as evidenced by stabilizing the mitochondrial membrane potential and reducing the overproduction of reactive oxygen species. Furthermore, hypoxia/reperfusion injury induced damaged mitochondrial axonal transport and consequently neurotransmitter release disorder, which were ameliorated by RD-1 treatment. Besides, RD-1 inhibited the downregulation of proteins related with mitochondrial transport and neurotransmitter release induced by ischemic injury both in vivo and in vitro. The obtained data demonstrated the neuroprotective effect of RD-1 and the involved mechanisms were partially attributed to the improvement in mitochondrial function and the synaptic activity. Our study indicated that RD-1 may be a potential therapeutic drug for the ischemic stroke therapy.
Collapse
|
39
|
Shah SA, Amin FU, Khan M, Abid MN, Rehman SU, Kim TH, Kim MW, Kim MO. Anthocyanins abrogate glutamate-induced AMPK activation, oxidative stress, neuroinflammation, and neurodegeneration in postnatal rat brain. J Neuroinflammation 2016; 13:286. [PMID: 27821173 PMCID: PMC5100309 DOI: 10.1186/s12974-016-0752-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/26/2016] [Indexed: 12/20/2022] Open
Abstract
Background Glutamate-induced excitotoxicity, oxidative damage, and neuroinflammation are believed to play an important role in the development of a number of CNS disorders. We recently reported that a high dose of glutamate could induce AMPK-mediated neurodegeneration in the postnatal day 7 (PND7) rat brain. Yet, the mechanism of glutamate-induced oxidative stress and neuroinflammation in the postnatal brain is not well understood. Here, we report for the first time the mechanism of glutamate-induced oxidative damage, neuroinflammation, and neuroprotection by polyphenolic anthocyanins in PND7. Methods PND7 rat brains, SH-SY5Y, and BV2 cells treated either alone with glutamate or in combination with anthocyanins and compound C were examined with Western blot and immunofluorescence techniques. Additionally, reactive oxygen species (ROS) assay and other ELISA kit assays were employed to know the therapeutic efficacy of anthocyanins against glutamate. Results A single injection of glutamate to developing rats significantly increased brain glutamate levels, activated and phosphorylated AMPK induction, and inhibited nuclear factor-E2-related factor 2 (Nrf2) after 2, 3, and 4 h in a time-dependent manner. In contrast, anthocyanin co-treatment significantly reduced glutamate-induced AMPK induction, ROS production, neuroinflammation, and neurodegeneration in the developing rat brain. Most importantly, anthocyanins increased glutathione (GSH and GSSG) levels and stimulated the endogenous antioxidant system, including Nrf2 and heme oxygenase-1 (HO-1), against glutamate-induced oxidative stress. Interestingly, blocking AMPK with compound C in young rats abolished glutamate-induced neurotoxicity. Similarly, all these experiments were replicated in SH-SY5Y cells by silencing AMPK with siRNA, which suggests that AMPK is the key mediator in glutamate-induced neurotoxicity. Conclusions Here, we report for the first time that anthocyanins can potentially decrease glutamate-induced neurotoxicity in young rats. Our work demonstrates that glutamate is toxic to the developing rat brain and that anthocyanins can minimize the severity of glutamate-induced neurotoxicity in an AMPK-dependent manner. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0752-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shahid Ali Shah
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Faiz Ul Amin
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Mehtab Khan
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Muhammad Noman Abid
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Shafiq Ur Rehman
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Tae Hyun Kim
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Min Woo Kim
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Myeong Ok Kim
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea.
| |
Collapse
|
40
|
Zhen X, Ng ESK, Lam FFY. Suppression of ischaemia-induced injuries in rat brain by protease-activated receptor-1 (PAR-1) activating peptide. Eur J Pharmacol 2016; 786:36-46. [DOI: 10.1016/j.ejphar.2016.05.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
|
41
|
The Cytomegalovirus protein pUL37×1 targets mitochondria to mediate neuroprotection. Sci Rep 2016; 6:31373. [PMID: 27562039 PMCID: PMC4999870 DOI: 10.1038/srep31373] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 07/19/2016] [Indexed: 12/22/2022] Open
Abstract
There is substantial evidence that mitochondrial dysfunction plays a significant role in the pathogenesis of Parkinson disease (PD). This contribution probably encompasses defects of oxidative phosphorylation, mitochondrial turnover (mitophagy), mitochondrial derived oxidative stress, and apoptotic signalling. Human cytomegalovirus immediate-early protein pUL37 × 1 induces Bax mitochondrial translocation and inactivation to prevent apoptosis. Over-expressing pUL37 × 1 in neuronal cells protects against staurosporin and 6-hydroxydopamine induced apoptosis and cell death. Protection is not enhanced by bax silencing in pUL37 × 1 over-expressing cells, suggesting a bax-dependent mechanism of action. pUL37 × 1 increases glycolysis and induces mitochondrial hyperpolarization, a bax independent anti-apoptotic action. pUL37 × 1 increases glycolysis through activation of phosphofructokinase by a calcium-dependent pathway. The dual anti-apoptotic mechanism of pUL37 × 1 may be considered a novel neuroprotective strategy in diseases where mitochondrial dysfunction and apoptotic pathways are involved.
Collapse
|
42
|
Lai SH, Li W, Yao JH, Han BJ, Jiang GB, Zhang C, Zeng CC, Liu YJ. Protein binding and anticancer activity studies of ruthenium(II) polypyridyl complexes toward BEL-7402 cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 158:39-48. [DOI: 10.1016/j.jphotobiol.2016.02.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 02/21/2016] [Accepted: 02/22/2016] [Indexed: 12/20/2022]
|
43
|
Tubastatin A, an HDAC6 inhibitor, alleviates stroke-induced brain infarction and functional deficits: potential roles of α-tubulin acetylation and FGF-21 up-regulation. Sci Rep 2016; 6:19626. [PMID: 26790818 PMCID: PMC4726180 DOI: 10.1038/srep19626] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/07/2015] [Indexed: 12/17/2022] Open
Abstract
Histone deacetylase (HDAC) 6 exists exclusively in cytoplasm and deacetylates cytoplasmic proteins such as α-tubulin. HDAC6 dysfunction is associated with several pathological conditions in the central nervous system. This study investigated the beneficial effects of tubastatin A (TubA), a novel specific HDAC6 inhibitor, in a rat model of transient middle cerebral artery occlusion (MCAO) and an in vitro model of excitotoxicity. Post-ischemic TubA treatment robustly improved functional outcomes, reduced brain infarction, and ameliorated neuronal cell death in MCAO rats. These beneficial effects lasted at least three days after MCAO. Notably, when given at 24 hours after MCAO, TubA still exhibited significant protection. Levels of acetylated α-tubulin were decreased in the ischemic hemisphere on Days 1 and 3 after MCAO, and were significantly restored by TubA. MCAO markedly downregulated fibroblast growth factor-21 (FGF-21) and TubA significantly reversed this downregulation. TubA also mitigated impaired FGF-21 signaling in the ischemic hemisphere, including up-regulating β-Klotho, and activating ERK and Akt/GSK-3β signaling pathways. In addition, both TubA and exogenous FGF-21 conferred neuroprotection and restored mitochondrial trafficking in rat cortical neurons against glutamate-induced excitotoxicity. Our findings suggest that the neuroprotective effects of TubA likely involve HDAC6 inhibition and the subsequent up-regulation of acetylated α-tubulin and FGF-21.
Collapse
|
44
|
Matic I, Cocco S, Ferraina C, Martin-Jimenez R, Florenzano F, Crosby J, Lupi R, Amadoro G, Russell C, Pignataro G, Annunziato L, Abramov AY, Campanella M. Neuroprotective coordination of cell mitophagy by the ATPase Inhibitory Factor 1. Pharmacol Res 2016; 103:56-68. [PMID: 26484591 DOI: 10.1016/j.phrs.2015.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 01/03/2023]
Abstract
The mitochondrial ATPase Inhibitory Factor 1 (hereafter referred to as IF1) blocks the reversal of the F1Fo-ATPsynthase to prevent detrimental consumption of cellular ATP and associated demise. Herein, we infer further its molecular physiology by assessing its protective function in neurons during conditions of challenged homeostatic respiration. By adopting in vitro and in vivo protocols of hypoxia/ischemia and re-oxygenation, we show that a shift in the IF1:F1Fo-ATPsynthase expression ratio occurs in neurons. This increased IF1 level is essential to induce accumulation of the PTEN-induced putative kinase 1 (PINK-1) and recruitment of the mitophagic ubiquitin ligase PARK-2 to promote autophagic "control" of the mitochondrial population. In IF1 overexpressing neurons ATP depletion is reduced during hypoxia/ischemia and the mitochondrial membrane potential (ΔYm) resilient to re-oxygenation as well as resistant to electrogenic, Ca(2+) dependent depolarization. These data suggest that in mammalian neurons mitochondria adapt to respiratory stress by upregulating IF1, which exerts a protective role by coordinating pro-survival cell mitophagy and bioenergetics resilience.
Collapse
Affiliation(s)
- Ivana Matic
- Department of Biology, University of Rome "TorVergata", 00133 Rome, Italy
| | - Stefania Cocco
- EBRI-European Brain Research Institute, 00143 Rome, Italy
| | - Caterina Ferraina
- Department of Biology, University of Rome "TorVergata", 00133 Rome, Italy; Regina Elena-National Cancer Institute, 00144 Rome, Italy
| | - Rebeca Martin-Jimenez
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street NW1 0TU, United Kingdom
| | | | - James Crosby
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street NW1 0TU, United Kingdom
| | - Ramona Lupi
- EBRI-European Brain Research Institute, 00143 Rome, Italy
| | - Giusy Amadoro
- EBRI-European Brain Research Institute, 00143 Rome, Italy
| | - Claire Russell
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street NW1 0TU, United Kingdom
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, School of Medicine, Federico II University of Naples, Italy; Department of Molecular Neuroscience, Institute of Neurology, University College London, United Kingdom
| | - Lucio Annunziato
- Division of Pharmacology, Department of Neuroscience, School of Medicine, Federico II University of Naples, Italy; Department of Molecular Neuroscience, Institute of Neurology, University College London, United Kingdom
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, Institute of Neurology, University College London, United Kingdom
| | - Michelangelo Campanella
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street NW1 0TU, United Kingdom; UCL Consortium for Mitochondrial Research, Royal College Street, University of London, United Kingdom; Department of Biology, University of Rome "TorVergata", 00133 Rome, Italy; Regina Elena-National Cancer Institute, 00144 Rome, Italy.
| |
Collapse
|
45
|
Resveratrol Protects PC12 Cell against 6-OHDA Damage via CXCR4 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:730121. [PMID: 26681969 PMCID: PMC4670657 DOI: 10.1155/2015/730121] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/21/2015] [Accepted: 10/25/2015] [Indexed: 12/20/2022]
Abstract
Resveratrol, herbal nonflavonoid polyphenolic compound naturally derived from grapes, has long been acknowledged to possess extensive biological and pharmacological properties including antioxidant and anti-inflammatory ones and may exert a neuroprotective effect on neuronal damage in neurodegenerative diseases. However, the underlying molecular mechanisms remain undefined. In the present study, we intended to investigate the neuroprotective effects of resveratrol against 6-OHDA-induced neurotoxicity of PC12 cells and further explore the possible mechanisms involved. For this purpose, PC12 cells were exposed to 6-OHDA in the presence of resveratrol (0, 12.5, 25, and 50 μM). The results showed that resveratrol increased cell viability, alleviated the MMP reduction, and reduced the number of apoptotic cells as measured by MTT assay, JC-1 staining, and Hoechst/PI double staining (all p < 0.01). Immunofluorescent staining and Western blotting revealed that resveratrol averts 6-OHDA induced CXCR4 upregulation (p < 0.01). Our results demonstrated that resveratrol could effectively protect PC12 cells from 6-OHDA-induced oxidative stress and apoptosis via CXCR4 signaling pathway.
Collapse
|
46
|
Mitochondrial Optic Atrophy (OPA) 1 Processing Is Altered in Response to Neonatal Hypoxic-Ischemic Brain Injury. Int J Mol Sci 2015; 16:22509-26. [PMID: 26393574 PMCID: PMC4613321 DOI: 10.3390/ijms160922509] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 08/26/2015] [Accepted: 09/11/2015] [Indexed: 01/02/2023] Open
Abstract
Perturbation of mitochondrial function and subsequent induction of cell death pathways are key hallmarks in neonatal hypoxic-ischemic (HI) injury, both in animal models and in term infants. Mitoprotective therapies therefore offer a new avenue for intervention for the babies who suffer life-long disabilities as a result of birth asphyxia. Here we show that after oxygen-glucose deprivation in primary neurons or in a mouse model of HI, mitochondrial protein homeostasis is altered, manifesting as a change in mitochondrial morphology and functional impairment. Furthermore we find that the mitochondrial fusion and cristae regulatory protein, OPA1, is aberrantly cleaved to shorter forms. OPA1 cleavage is normally regulated by a balanced action of the proteases Yme1L and Oma1. However, in primary neurons or after HI in vivo, protein expression of YmelL is also reduced, whereas no change is observed in Oma1 expression. Our data strongly suggest that alterations in mitochondria-shaping proteins are an early event in the pathogenesis of neonatal HI injury.
Collapse
|
47
|
Hong F, Zhao X, Chen M, Zhou Y, Ze Y, Wang L, Wang Y, Ge Y, Zhang Q, Ye L. TiO2 nanoparticles-induced apoptosis of primary cultured Sertoli cells of mice. J Biomed Mater Res A 2015; 104:124-35. [PMID: 26238530 DOI: 10.1002/jbm.a.35548] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/09/2015] [Accepted: 07/31/2015] [Indexed: 11/08/2022]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs), as largest production and use of nanomaterials, have been demonstrated to have a potential toxicity on reproductive system. However, the mechanism underlying male reproductive toxicity of TiO2 NPs remains limited. Thus, our study was designed to examine the cellular viability, apoptosis, oxidative stress, antioxidant capacity, and expression of apoptotic cytokines in primary cultured Sertoli cells isolated from mice under TiO2 NPs exposure. Results showed that TiO2 NPs exposure from 5 to 30 μg/mL resulted in reduction of cell viability, lactate dehydrogenase release, and induction of apoptosis or death on Sertoli cells. TiO2 NPs could migrate to Sertoli cells, which induced mitochondria-mediated or endoplasmic-reticulum-mediated apoptotic changes including elevation in reactive oxygen species (ROS) generation and reductions in superoxide dismutase, catalase, and glutathione peroxidase activities, decreases in mitochondrial membrane potential (ΔΨm), and releases of cytochrome c into the cytosol. In addition, upregulation of cytochrome c, Bax, caspase-3, glucose-regulated protein 78, and C/EBP homologous protein and caspase-12 protein expression, and downregulation of bcl-2 protein expression in primary cultured Sertoli cells induced by TiO2 NPs treatment. All of the results suggested that ROS generation may play a critical role in the initiation of TiO2 NPs-induced apoptosis by mediation of the disruption of ΔΨm, the cytochrome c release, and further the activation of caspase cascade and unfolded protein response signaling pathway.
Collapse
Affiliation(s)
- Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, People's Republic of China.,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, People's Republic of China.,School of Life Sciences, Huaiyin Normal University, Huaian, People's Republic of China
| | - Xiaoyang Zhao
- Medical College of Soochow University, Suzhou, People's Republic of China
| | - Ming Chen
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, People's Republic of China.,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, People's Republic of China.,School of Life Sciences, Huaiyin Normal University, Huaian, People's Republic of China
| | - Yingjun Zhou
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, People's Republic of China.,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, People's Republic of China.,School of Life Sciences, Huaiyin Normal University, Huaian, People's Republic of China
| | - Yuguan Ze
- Medical College of Soochow University, Suzhou, People's Republic of China
| | - Ling Wang
- Library of Soochow University, Suzhou, People's Republic of China
| | - Yajing Wang
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, People's Republic of China.,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, People's Republic of China.,School of Life Sciences, Huaiyin Normal University, Huaian, People's Republic of China
| | - Yushuang Ge
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, People's Republic of China.,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, People's Republic of China.,School of Life Sciences, Huaiyin Normal University, Huaian, People's Republic of China
| | - Qi Zhang
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, People's Republic of China.,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, People's Republic of China.,School of Life Sciences, Huaiyin Normal University, Huaian, People's Republic of China
| | - Lingqun Ye
- Medical College of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
48
|
Xu SY, Hu FY, Ren LJ, Chen L, Zhou ZQ, Zhang XJ, Li WP. Dantrolene enhances the protective effect of hypothermia on cerebral cortex neurons. Neural Regen Res 2015; 10:1279-85. [PMID: 26487856 PMCID: PMC4590241 DOI: 10.4103/1673-5374.162761] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2015] [Indexed: 01/05/2023] Open
Abstract
Therapeutic hypothermia is the most promising non-pharmacological neuroprotective strategy against ischemic injury. However, shivering is the most common adverse reaction. Many studies have shown that dantrolene is neuroprotective in in vitro and in vivo ischemic injury models. In addition to its neuroprotective effect, dantrolene neutralizes the adverse reaction of hypothermia. Dantrolene may be an effective adjunctive therapy to enhance the neuroprotection of hypothermia in treating ischemic stroke. Cortical neurons isolated from rat fetuses were exposed to 90 minutes of oxygen-glucose deprivation followed by reoxygenation. Neurons were treated with 40 μM dantrolene, hypothermia (at 33°C), or the combination of both for 12 hours. Results revealed that the combination of dantrolene and hypothermia increased neuronal survival and the mitochondrial membrane potential, and reduced intracellular active oxygen cytoplasmic histone-associated DNA fragmentation, and apoptosis. Furthermore, improvements in cell morphology were observed. The combined treatment enhanced these responses compared with either treatment alone. These findings indicate that dantrolene may be used as an effective adjunctive therapy to enhance the neuroprotective effects of hypothermia in ischemic stroke.
Collapse
Affiliation(s)
- Sui-yi Xu
- Postdoctoral Workstation, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Feng-yun Hu
- Department of Neurology, Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li-jie Ren
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Lei Chen
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Zhu-qing Zhou
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Xie-jun Zhang
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Wei-ping Li
- Postdoctoral Workstation, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| |
Collapse
|
49
|
Mitochondrial CB1 receptor is involved in ACEA-induced protective effects on neurons and mitochondrial functions. Sci Rep 2015. [PMID: 26215450 PMCID: PMC4516969 DOI: 10.1038/srep12440] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Mitochondrial dysfunction contributes to cell death after cerebral ischemia/reperfusion (I/R) injury. Cannabinoid CB1 receptor is expressed in neuronal mitochondrial membranes (mtCB1R) and involved in regulating mitochondrial functions under physiological conditions. However, whether mtCB1R affords neuroprotection against I/R injury remains unknown. We used mouse models of cerebral I/R, primary cultured hippocampal neurons exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) and Ca(2+)-induced injury in purified neuronal mitochondria to investigate the role of mtCB1R in neuroprotection. Our results showed selective cell-permeant CB1 receptor agonist, arachidonyl-2-chloroethylamide (ACEA), significantly up-regulated the expression of mtCB1R protein in hippocampal neurons and tissue. In vitro, ACEA restored cell viability, inhibited generation of reactive oxygen species (ROS), decreased lactate dehydrogenase (LDH) release and reduced apoptosis, improved mitochondrial function. In vivo, ACEA ameliorated neurological scores, diminished the number of TUNEL-positive neurons and decreased the expression of cleaved caspase-3. However, ACEA-induced benefits were blocked by the selective cell-permeant CB1 receptor antagonist AM251, but just partially by the selective cell-impermeant CB1 receptor antagonist hemopressin. In purified neuronal mitochondria, mtCB1R activation attenuated Ca(2+)-induced mitochondrial injury. In conclusion, mtCB1R is involved in ACEA-induced protective effects on neurons and mitochondrial functions, suggesting mtCB1R may be a potential novel target for the treatment of brain ischemic injury.
Collapse
|
50
|
Yang W, Li L, Huang R, Pei Z, Liao S, Zeng J. Hypoxia inducible factor-1alpha mediates protection of DL-3-n-butylphthalide in brain microvascular endothelial cells against oxygen glucose deprivation-induced injury. Neural Regen Res 2015; 7:948-54. [PMID: 25722681 PMCID: PMC4341293 DOI: 10.3969/j.issn.1673-5374.2012.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 03/28/2012] [Indexed: 02/03/2023] Open
Abstract
Studies have demonstrated that DL-3-n-butylphthalide can significantly alleviate oxygen glucose deprivation-induced injury of human umbilical vein endothelial cells at least partly associated with its enhancement on oxygen glucose deprivation -induced hypoxia inducible factor-1α expression. In this study, we hypothesized that DL-3-n-butylphthalide can protect against oxygen glucose deprivation-induced injury of newborn rat brain microvascular endothelial cells by means of upregulating hypoxia inducible factor-1α expression. MTT assay and Hoechst staining results showed that DL-3-n-butylphthalide protected brain microvascular endothelial cells against oxygen glucose deprivation-induced injury in a dose-dependent manner. Western blot and immunofluorescent staining results further confirmed that the protective effect was related to upregulation of hypoxia inducible factor-1α. Real-time RT-PCR reaction results showed that DL-3-n-butylphthalide reduced apoptosis by inhibiting downregulation of pro-apoptotic gene caspase-3 mRNA expression and upregulation of apoptosis-executive protease bcl-2 mRNA expression; however, DL-3-n-butylphthalide had no protective effects on brain microvascular endothelial cells after knockdown of hypoxia inducible factor-1α by small interfering RNA. These findings suggest that DL-3-n-butylphthalide can protect brain microvascular endothelial cells against oxygen glucose deprivation-induced injury by upregulating bcl-2 expression and downregulating caspase-3 expression though hypoxia inducible factor-1α pathway.
Collapse
Affiliation(s)
- Weihong Yang
- Department of Neurology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Ling Li
- Department of Neurology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Ruxun Huang
- Department of Neurology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Zhong Pei
- Department of Neurology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Songjie Liao
- Department of Neurology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Jinsheng Zeng
- Department of Neurology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| |
Collapse
|