1
|
Li Q, Song Z, Peng L, Feng S, Zhan K, Ling H. Dihydromyricetin Improves High Glucose-Induced Dopaminergic Neuronal Damage by Activating AMPK-Autophagy Signaling Pathway. Exp Clin Endocrinol Diabetes 2024; 132:631-641. [PMID: 39168148 DOI: 10.1055/a-2399-1174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
INTRODUCTION In recent years, a growing number of clinical and biological studies have shown that patients with type 2 diabetes mellitus (T2DM) are at increased risk of developing Parkinson's disease (PD). Prolonged exposure to hyperglycemia results in abnormal glucose metabolism, which in turn causes pathological changes similar to PD, leading to selective loss of dopaminergic neurons in the compact part of the substantia nigra. Dihydromyricetin (DHM) is a naturally occurring flavonoid with various biological activities including antioxidant and hepatoprotective properties. In this study, the effect of DHM on high glucose-induced dopaminergic neuronal damage was investigated. METHODS The potential modulatory effects of DHM on high glucose-induced dopaminergic neuronal damage and its mechanism were studied. RESULTS DHM ameliorated high glucose-induced dopaminergic neuronal damage and autophagy injury. Inhibition of autophagy by 3-methyladenine abrogated the beneficial effects of DHM on high glucose-induced dopaminergic neuronal damage. In addition, DHM increased levels of p-AMP-activated protein kinase (AMPK) and phosphorylated UNC51-like kinase 1. The AMPK inhibitor compound C eliminated DHM-induced autophagy and subsequently inhibited the ameliorative effects of DHM on high glucose-induced dopaminergic neuronal damage. DISCUSSION DHM ameliorates high glucose-induced dopaminergic neuronal damage by activating the AMPK-autophagy pathway.
Collapse
Affiliation(s)
- Qi Li
- Department of Physiology, University of South China Hengyang Medical School, Hengyang, China
- Department of Pathology, Yuebei People's Hospital Affiliated to Shantou University School of Medicine, Shaoguan, China
| | - Zhenjiang Song
- Department of Physiology, University of South China Hengyang Medical School, Hengyang, China
| | - Liting Peng
- Department of Physiology, University of South China Hengyang Medical School, Hengyang, China
| | - Shuidong Feng
- Department of Social Medicine and Health Management, University of South China Hengyang Medical School, Hengyang, China
| | - Kebin Zhan
- Department of Neurology, The Second Affiliated Hospital, University of South China Hengyang Medical School, Hengyang, China
| | - Hongyan Ling
- Department of Physiology, University of South China Hengyang Medical School, Hengyang, China
| |
Collapse
|
2
|
Gillane R, Daygon D, Khalil ZG, Marcellin E. Biosynthesis of novel non-proteinogenic amino acids β-hydroxyenduracididine and β-methylphenylalanine in Escherichia coli. Front Bioeng Biotechnol 2024; 12:1468974. [PMID: 39444519 PMCID: PMC11496134 DOI: 10.3389/fbioe.2024.1468974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Non-proteinogenic amino acids (npAAs) are valuable building blocks for the development of advanced pharmaceuticals and agrochemicals. The surge in interest in their synthesis is primarily due to the potential to enhance and diversify existing bioactive molecules. This can be achieved by altering these bioactive molecules to improve their effectiveness, reducing resistance compared to their natural counterparts or generating molecules with novel functions. Traditional production of npAAs in native hosts requires specialized conditions and complex cultivation media. Furthermore, these compounds are often found in organisms that challenge genetic manipulation. Thus, the recombinant production of these npAAs in a model organism like Escherichia coli paves the way for groundbreaking advancements in synthetic biology. Two synthetic operons, comprising of five heterologous proteins were genomically integrated into E. coli for the synthesis of npAAs β-methylphenylalanine (BmePhe), β-hydroxyenduracididine (BhEnd), and enduracididine (End). Proteomic and metabolomic analysis confirmed production of these compounds in E. coli for the first time. Interestingly, we discovered that the exogenous addition of pathway precursors to the E. coli system enhanced the yield of BmePhe by 2.5 times, whereas it concurrently attenuated the production of BhEnd and End, signifying a selective precursor-dependent yield enhancement. The synthetic biology landscape is broadened in this study by expanding the repertoire of amino acids beyond the conventional set of 22 standard proteinogenic amino acids. The biosynthesized npAAs, End, BhEnd, and BmePhe hold promise for engineering proteins with modified functions, integrating into novel metabolites and/or enhancing biological stability and activity. Additionally, these amino acids' biological production and subsequent purification present an alternative to traditional chemical synthesis methods, paving a direct pathway for pharmacological evaluation.
Collapse
Affiliation(s)
- Rosemary Gillane
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
- ARC Centre of Excellence in Synthetic Biology, University of Queensland, Brisbane, QLD, Australia
| | - Dara Daygon
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
- Queensland Metabolomics and Proteomics Facility, University of Queensland, Brisbane, QLD, Australia
| | - Zeinab G. Khalil
- Institute of Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Esteban Marcellin
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
- ARC Centre of Excellence in Synthetic Biology, University of Queensland, Brisbane, QLD, Australia
- Queensland Metabolomics and Proteomics Facility, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
3
|
Huang L, Lin R, Zhang C, Zheng S, Wang Y, Wu Z, Chen S, Shen Y, Zhang G, Qi Y, Lin L. The Neuroprotective and Anxiolytic Effects of Magnesium Sulfate on Retinal Dopaminergic Neurons in 6-OHDA-Induced Parkinsonian Rats: A Pilot Study. Brain Sci 2024; 14:861. [PMID: 39335357 PMCID: PMC11430011 DOI: 10.3390/brainsci14090861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
This study investigates the protective effects of magnesium sulfate on dopamine neurons in the retinas of rats with 6-hydroxydopamine (6-OHDA)-induced Parkinson's disease (PD). Rapidly progressing cognitive decline often precedes or coincides with the motor symptoms associated with PD. PD patients also frequently exhibit visual function abnormalities. However, the specific mechanisms underlying visual dysfunction in PD patients are not yet fully understood. Therefore, this study aims to investigate whether magnesium homeostasis affects dopaminergic neurons in the retina of PD rats. Thirty-six rats were divided into four groups: (1) control, (2) control with magnesium sulfate (control/MgSO4), (3) Parkinson's disease (PD), and (4) Parkinson's disease with magnesium sulfate (PD/MgSO4). The apomorphine-induced (APO) rotation test assessed the success of the PD models. The open-field experiment measured the rats' anxiety levels. Tyrosine hydroxylase (TH) and glutamate levels, indicators of dopamine neuron survival, were detected using immunofluorescence staining. Protein levels of solute carrier family 41 A1 (SCL41A1), magnesium transporter 1 (MagT1), and cyclin M2 (CNNM2) in the retina were analyzed using Western blot. Results showed that, compared to the PD group, rats in the PD/MgSO4 group had improved psychological states and motor performance at two and four weeks post-surgery. The PD/MgSO4 group also exhibited significantly higher TH fluorescence intensity in the left retinas and lower glutamate fluorescence intensity than the PD group. Additional experiments indicated that the protein levels of SLC41A1, MagT1, and CNNM2 were generally higher in the retinas of the PD/MgSO4 group, along with an increase in retinal magnesium ion content. This suggests that magnesium sulfate may reduce glutamate levels and protect dopamine neurons in the retina. Thus, magnesium sulfate might have therapeutic potential for visual functional impairments in PD patients.
Collapse
Affiliation(s)
- Leyi Huang
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
- Key Laboratory of Brain Aging and Neurodegenerative Disease, Fujian Medical University, Fuzhou 350122, China
| | - Renxi Lin
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
- Key Laboratory of Brain Aging and Neurodegenerative Disease, Fujian Medical University, Fuzhou 350122, China
- Experimental Teaching Center of Basic Medicine, Fujian Medical University, Fuzhou 350122, China
| | - Chunying Zhang
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
| | - Shaoqing Zheng
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Yiyang Wang
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Zeyu Wu
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Sihao Chen
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Yihan Shen
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China; (S.Z.); (Y.W.); (Z.W.); (S.C.); (Y.S.)
| | - Guoheng Zhang
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
| | - Yuanlin Qi
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
| | - Ling Lin
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350122, China; (L.H.); (R.L.); (C.Z.); (G.Z.); (Y.Q.)
- Key Laboratory of Brain Aging and Neurodegenerative Disease, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
4
|
Javid H, Rahimian R, Salimi M, Haghani-Samani E, Farhadi M, Torkaman-Boutorabi A. Fumaria vaillantii extract protects PC12 cells against neurotoxicity induced by 6-OHDA. Mol Biol Rep 2024; 51:768. [PMID: 38884894 DOI: 10.1007/s11033-024-09673-5] [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: 01/09/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Parkinson's disease is a neurological disorder caused by the loss of dopaminergic neurons in the midbrain. Various mechanisms are involved in the incidence of the disease including oxidative stress. Several herbs and natural products may interfere with the oxidative-stress pathway due to their antioxidant effects. OBJECTIVE Herein, we aimed to investigate the neuroprotective role of F. vaillantii extract on Parkinson's in vitro and in vivo model owing to the presence of the bioactive agents with antioxidant properties. METHODS In vitro experments showed that 6-hydroxydopamine could induce toxicity in PC12 cells. The impact of F. vaillantii extract on cell viability was measured by using MTT assay. Nuclear morphological changes were qualitatively evaluated employing Hoechst staining. The antioxidant activity of the extract was determined by ROS and lipid peroxidation assays. Tyrosine hydroxylase protein expression was measured by western blotting in PC12 cells. For in vivo study, movement parameters were evaluated. RESULTS The results indicated that 75 µΜ of 6-OHDA induced 50% toxicity in PC12 cells for 24 h. Following post-treatment with F. vaillantii extract (0.1 mg/ml) for 72 h, we observed that the extract effectively prevented cell toxicity induced by 6-OHDA and reduced the apoptotic cell population. Furthermore, the extract attenuated the ROS level, lipid peroxidation and increased protein expression of TH after 72 h of treatment. In addition, oral administration of 300 mg/kg of F. vaillantii extract for 14 days improved locomotor activity, catalepsy, bradykinesia, motor coordination and reduced the apomorphine-caused rotation in 6-OHDA- induced Parkinson's disease-like symptoms in male rats. CONCLUSION The present study suggests a protective role for the extract of F. vaillantii against oxidative stress-induced cell damage in the PC12 cells exposed to neurotoxin 6-OHDA which was verified in in vivo model by reducing the motor defects induced by 6-OHDA. This extract could be a promising therapeutic agent for the prevention of PD progression.
Collapse
Affiliation(s)
- Hanieh Javid
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, No. 88, Italya Street, Vesaal Shirazi Avenue, Keshavars Boulevard, Tehran, Iran
| | - Rana Rahimian
- Department of Microbiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Misha Salimi
- Department of Biology, Faculty of Converging Sciences and Technology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Elaheh Haghani-Samani
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, No. 88, Italya Street, Vesaal Shirazi Avenue, Keshavars Boulevard, Tehran, Iran
| | - Mona Farhadi
- Department of Microbiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Anahita Torkaman-Boutorabi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, No. 88, Italya Street, Vesaal Shirazi Avenue, Keshavars Boulevard, Tehran, Iran.
- Research Center for Cognitive and Behavioral Studies, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Timón-Gómez A, Scharr AL, Wong NY, Ni E, Roy A, Liu M, Chau J, Lampert JL, Hireed H, Kim NS, Jan M, Gupta AR, Day RW, Gardner JM, Wilson RJA, Barrientos A, Chang AJ. Tissue-specific mitochondrial HIGD1C promotes oxygen sensitivity in carotid body chemoreceptors. eLife 2022; 11:e78915. [PMID: 36255054 PMCID: PMC9635879 DOI: 10.7554/elife.78915] [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: 03/24/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Mammalian carotid body arterial chemoreceptors function as an early warning system for hypoxia, triggering acute life-saving arousal and cardiorespiratory reflexes. To serve this role, carotid body glomus cells are highly sensitive to decreases in oxygen availability. While the mitochondria and plasma membrane signaling proteins have been implicated in oxygen sensing by glomus cells, the mechanism underlying their mitochondrial sensitivity to hypoxia compared to other cells is unknown. Here, we identify HIGD1C, a novel hypoxia-inducible gene domain factor isoform, as an electron transport chain complex IV-interacting protein that is almost exclusively expressed in the carotid body and is therefore not generally necessary for mitochondrial function. Importantly, HIGD1C is required for carotid body oxygen sensing and enhances complex IV sensitivity to hypoxia. Thus, we propose that HIGD1C promotes exquisite oxygen sensing by the carotid body, illustrating how specialized mitochondria can be used as sentinels of metabolic stress to elicit essential adaptive behaviors.
Collapse
Affiliation(s)
| | - Alexandra L Scharr
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Nicholas Y Wong
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Erwin Ni
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Arijit Roy
- Department of Physiology and Pharmacology, University of CalgaryCalgaryCanada
- Hotchkiss Brain Institute, University of CalgaryCalgaryCanada
- Alberta Children's Hospital Research Institute, University of CalgaryCalgaryCanada
| | - Min Liu
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Julisia Chau
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Jack L Lampert
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Homza Hireed
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Noah S Kim
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Masood Jan
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Alexander R Gupta
- Department of Surgery, University of California, San FranciscoSan FranciscoUnited States
- Diabetes Center, University of California, San FranciscoSan FranciscoUnited States
| | - Ryan W Day
- Department of Surgery, University of California, San FranciscoSan FranciscoUnited States
| | - James M Gardner
- Department of Surgery, University of California, San FranciscoSan FranciscoUnited States
- Diabetes Center, University of California, San FranciscoSan FranciscoUnited States
| | - Richard JA Wilson
- Department of Physiology and Pharmacology, University of CalgaryCalgaryCanada
- Hotchkiss Brain Institute, University of CalgaryCalgaryCanada
- Alberta Children's Hospital Research Institute, University of CalgaryCalgaryCanada
| | | | - Andy J Chang
- Department of Physiology and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
6
|
Wang PP, Song X, Zhao XK, Wei MX, Gao SG, Zhou FY, Han XN, Xu RH, Wang R, Fan ZM, Ren JL, Li XM, Wang XZ, Yang MM, Hu JF, Zhong K, Lei LL, Li LY, Chen Y, Chen YJ, Ji JJ, Yang YZ, Li J, Wang LD. Serum Metabolomic Profiling Reveals Biomarkers for Early Detection and Prognosis of Esophageal Squamous Cell Carcinoma. Front Oncol 2022; 12:790933. [PMID: 35155234 PMCID: PMC8832491 DOI: 10.3389/fonc.2022.790933] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/04/2022] [Indexed: 11/15/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common aggressive malignancies worldwide, particularly in northern China. The absence of specific early symptoms and biomarkers leads to late-stage diagnosis, while early diagnosis and risk stratification are crucial for improving overall prognosis. We performed UPLC-MS/MS on 450 ESCC patients and 588 controls consisting of a discovery group and two validation groups to identify biomarkers for early detection and prognosis. Bioinformatics and clinical statistical methods were used for profiling metabolites and evaluating potential biomarkers. A total of 105 differential metabolites were identified as reliable biomarker candidates for ESCC with the same tendency in three cohorts, mainly including amino acids and fatty acyls. A predictive model of 15 metabolites [all-trans-13,14-dihydroretinol, (±)-myristylcarnitine, (2S,3S)-3-methylphenylalanine, 3-(pyrazol-1-yl)-L-alanine, carnitine C10:1, carnitine C10:1 isomer1, carnitine C14-OH, carnitine C16:2-OH, carnitine C9:1, formononetin, hyodeoxycholic acid, indole-3-carboxylic acid, PysoPE 20:3, PysoPE 20:3(2n isomer1), and resolvin E1] was developed by logistic regression after LASSO and random forest analysis. This model held high predictive accuracies on distinguishing ESCC from controls in the discovery and validation groups (accuracies > 89%). In addition, the levels of four downregulated metabolites [hyodeoxycholic acid, (2S,3S)-3-methylphenylalanine, carnitine C9:1, and indole-3-carboxylic acid] were significantly higher in early cancer than advanced cancer. Furthermore, three independent prognostic markers were identified by multivariate Cox regression analyses with and without clinical indicators: a high level of MG(20:4)isomer and low levels of 9,12-octadecadienoic acid and L-isoleucine correlated with an unfavorable prognosis; the risk score based on these three metabolites was able to stratify patients into low or high risk. Moreover, pathway analysis indicated that retinol metabolism and linoleic acid metabolism were prominent perturbed pathways in ESCC. In conclusion, metabolic profiling revealed that perturbed amino acids and lipid metabolism were crucial metabolic signatures of ESCC. Both panels of diagnostic and prognostic markers showed excellent predictive performances. Targeting retinol and linoleic acid metabolism pathways may be new promising mechanism-based therapeutic approaches. Thus, this study would provide novel insights for the early detection and risk stratification for the clinical management of ESCC and potentially improve the outcomes of ESCC.
Collapse
Affiliation(s)
- Pan Pan Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xin Song
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xue Ke Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Meng Xia Wei
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - She Gan Gao
- Department of Oncology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Fu You Zhou
- Department of Thoracic Surgery, Anyang Tumor Hospital, Anyang, China
| | - Xue Na Han
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Rui Hua Xu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ran Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Zong Min Fan
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Jing Li Ren
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue Min Li
- Department of Pathology, Hebei Provincial Cixian People’s Hospital, Cixian, China
| | - Xian Zeng Wang
- Department of Thoracic Surgery, Linzhou People’s Hospital, Linzhou, China
| | - Miao Miao Yang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Jing Feng Hu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Kan Zhong
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ling Ling Lei
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Liu Yu Li
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yao Chen
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ya Jie Chen
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Jia Jia Ji
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yuan Ze Yang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Jia Li
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Li Dong Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- *Correspondence: Li Dong Wang,
| |
Collapse
|
7
|
Neurotoxicity and underlying cellular changes of 21 mitochondrial respiratory chain inhibitors. Arch Toxicol 2021; 95:591-615. [PMID: 33512557 PMCID: PMC7870626 DOI: 10.1007/s00204-020-02970-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022]
Abstract
Inhibition of complex I of the mitochondrial respiratory chain (cI) by rotenone and methyl-phenylpyridinium (MPP +) leads to the degeneration of dopaminergic neurons in man and rodents. To formally describe this mechanism of toxicity, an adverse outcome pathway (AOP:3) has been developed that implies that any inhibitor of cI, or possibly of other parts of the respiratory chain, would have the potential to trigger parkinsonian motor deficits. We used here 21 pesticides, all of which are described in the literature as mitochondrial inhibitors, to study the general applicability of AOP:3 or of in vitro assays that are assessing its activation. Five cI, three complex II (cII), and five complex III (cIII) inhibitors were characterized in detail in human dopaminergic neuronal cell cultures. The NeuriTox assay, examining neurite damage in LUHMES cells, was used as in vitro proxy of the adverse outcome (AO), i.e., of dopaminergic neurodegeneration. This test provided data on whether test compounds were unspecific cytotoxicants or specifically neurotoxic, and it yielded potency data with respect to neurite degeneration. The pesticide panel was also examined in assays for the sequential key events (KE) leading to the AO, i.e., mitochondrial respiratory chain inhibition, mitochondrial dysfunction, and disturbed proteostasis. Data from KE assays were compared to the NeuriTox data (AO). The cII-inhibitory pesticides tested here did not appear to trigger the AOP:3 at all. Some of the cI/cIII inhibitors showed a consistent AOP activation response in all assays, while others did not. In general, there was a clear hierarchy of assay sensitivity: changes of gene expression (biomarker of neuronal stress) correlated well with NeuriTox data; mitochondrial failure (measured both by a mitochondrial membrane potential-sensitive dye and a respirometric assay) was about 10–260 times more sensitive than neurite damage (AO); cI/cIII activity was sometimes affected at > 1000 times lower concentrations than the neurites. These data suggest that the use of AOP:3 for hazard assessment has a number of caveats: (i) specific parkinsonian neurodegeneration cannot be easily predicted from assays of mitochondrial dysfunction; (ii) deriving a point-of-departure for risk assessment from early KE assays may overestimate toxicant potency. Comparison of 21 data-rich mitochondrial toxicants for neurotoxicity Quantitative comparison of key event triggering thresholds for AOP:3 Comparison of two cell models and two exposure times for neurotoxicity Comparison of transcriptome changes and classical key event measures for sensitivity
Collapse
|
8
|
Feng Y, Ma J, Yuan L. β-Methylphenylalanine exerts neuroprotective effects in a Parkinson's disease model by protecting against tyrosine hydroxylase depletion. J Cell Mol Med 2020; 24:9871-9880. [PMID: 32697044 PMCID: PMC7520294 DOI: 10.1111/jcmm.15571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/03/2020] [Accepted: 06/10/2020] [Indexed: 01/27/2023] Open
Abstract
We evaluated the neuroprotective effects of β-methylphenylalanine in an experimental model of rotenone-induced Parkinson's disease (PD) in SH-SY5Y cells and rats. Cells were pre-treated with rotenone (2.5 µg/mL) for 24 hours followed by β-methylphenylalanine (1, 10 and 100 mg/L) for 72 hours. Cell viability, reactive oxygen species (ROS) levels, mitochondrial membrane potential (MMP), mitochondrial fragmentation, apoptosis, and mRNA and protein levels of tyrosine hydroxylase were determined. In a rat model of PD, dopamine (DA) and 3,4-dihydroxyphenylacetic acid (DOPAC) levels, bradykinesia and tyrosine hydroxylase expression were determined. In rotenone-pre-treated cells, β-methylphenylalanine significantly increased cell viability and MMP, whereas ROS levels, apoptosis and fragmented mitochondria were reduced. β-Methylphenylalanine significantly increased the mRNA and protein levels of tyrosine hydroxylase in SH-SY5Y cells. In the rotenone-induced rat model of PD, oral administration of β-methylphenylalanine recovered DA and DOPAC levels and bradykinesia. β-Methylphenylalanine significantly increased the protein expression of tyrosine hydroxylase in the striatum and substantia nigra of rats. In addition, in silico molecular docking confirmed binding between tyrosine hydroxylase and β-methylphenylalanine. Our experimental results show neuroprotective effects of β-methylphenylalanine via the recovery of mitochondrial damage and protection against the depletion of tyrosine hydroxylase. We propose that β-methylphenylalanine may be useful in the treatment of PD.
Collapse
Affiliation(s)
- Yan Feng
- Department of NeurologyHenan Provincial People's HospitalZhengzhouChina
| | - Jianjun Ma
- Department of NeurologyHenan Provincial People's HospitalZhengzhouChina
| | - Lipin Yuan
- Department of NeurologyHenan Provincial People's HospitalZhengzhouChina
| |
Collapse
|