1
|
Fu DX, Lei YT, Guo HB, Chen T, Gao XY, Wang XL, Huang X, Song LL, Wang SY, Dai QX. PRDX1 affects acrylamide-induced neural damage through the PTEN/AKT signaling pathway. Neurotoxicology 2025; 108:150-158. [PMID: 40189060 DOI: 10.1016/j.neuro.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Peroxiredoxin 1 (PRDX1) is a member of the peroxidase family of antioxidant enzymes. However, the role and mechanism of PRDX1 in acrylamide (ACR)-induced nerve damage have not been reported. We used SD rats and well-differentiated rat pheochromocytoma cells (PC-12 cells) to established in vivo and in vitro models of ACR. Immunohistochemistry, immunofluorescence and RT-qPCR experiments were used to detect the expression of PRDX1 in neurons of rat hippocampal tissue. The ultrastructural changes of neurons and PC-12 cells in rat hippocampal tissue were observed under transmission electron microscope. Western blot detected the protein expression levels of PRDX1, PTEN, AKT and p-AKT. In vivo and in vitro experimental results showed that PRDX1 showed a significant up-regulation trend after ACR exposure (p < 0.05). In vitro experiments showed that after inhibiting PRDX1 expression with PRDX1 siRNA, the survival rate of PC-12 cells significantly increased, and the damage to cell morphology and organelles was markedly improved. Western blot analysis revealed that ACR exposure can cause a significant increase in PTEN protein expression level and p-AKT/AKT protein ratio (p < 0.05). After inhibiting the expression of PRDX1, the protein expression level of PTEN and the protein ratio of p-AKT/AKT were significantly reduced, while the protein levels of SYN1 and BDNF were significantly increased (p < 0.05). This study, for the first time, demonstrates that PRDX1 affects ACR-induced neurotoxicity by regulating the PTEN/AKT signaling pathway. And, provides novel insights into the prevention and treatment of neurotoxicity in populations exposed to ACR.
Collapse
Affiliation(s)
- Dong-Xue Fu
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China
| | - Ya-Ting Lei
- Department of Occupational Health, Public Health College, Harbin Medical University, Harbin, PR China; Department of Preventive Medicine, Department of medicine, Qiannan medical college for nationalities, Guizhou Province, PR China
| | - Hai-Bo Guo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China
| | - Ting Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China
| | - Xiang-Ying Gao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China
| | - Xiao-Li Wang
- Department of Occupational Health, Public Health College, Harbin Medical University, Harbin, PR China
| | | | - Ling-Ling Song
- Department of Neurology, The Second Hospital of Chaoyang (Women's and Children's Hospital of Chaoyang), PR China.
| | - Sheng-Yuan Wang
- Department of Occupational Health, Public Health College, Harbin Medical University, Harbin, PR China.
| | - Qin-Xue Dai
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, PR China.
| |
Collapse
|
2
|
Jang E, Yu H, Kim E, Hwang J, Yoo J, Choi J, Jeong HS, Jang S. The Therapeutic Effects of Blueberry-Treated Stem Cell-Derived Extracellular Vesicles in Ischemic Stroke. Int J Mol Sci 2024; 25:6362. [PMID: 38928069 PMCID: PMC11203670 DOI: 10.3390/ijms25126362] [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: 04/15/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
An ischemic stroke, one of the leading causes of morbidity and mortality, is caused by ischemia and hemorrhage resulting in impeded blood supply to the brain. According to many studies, blueberries have been shown to have a therapeutic effect in a variety of diseases. Therefore, in this study, we investigated whether blueberry-treated mesenchymal stem cell (MSC)-derived extracellular vesicles (B-EVs) have therapeutic effects in in vitro and in vivo stroke models. We isolated the extracellular vesicles using cryo-TEM and characterized the particles and concentrations using NTA. MSC-derived extracellular vesicles (A-EVs) and B-EVs were round with a lipid bilayer structure and a diameter of ~150 nm. In addition, A-EVs and B-EVs were shown to affect angiogenesis, cell cycle, differentiation, DNA repair, inflammation, and neurogenesis following KEGG pathway and GO analyses. We investigated the protective effects of A-EVs and B-EVs against neuronal cell death in oxygen-glucose deprivation (OGD) cells and a middle cerebral artery occlusion (MCAo) animal model. The results showed that the cell viability was increased with EV treatment in HT22 cells. In the animal, the size of the cerebral infarction was decreased, and the behavioral assessment was improved with EV injections. The levels of NeuN and neurofilament heavy chain (NFH)-positive cells were also increased with EV treatment yet decreased in the MCAo group. In addition, the number of apoptotic cells was decreased with EV treatment compared with ischemic animals following TUNEL and Bax/Bcl-2 staining. These data suggested that EVs, especially B-EVs, had a therapeutic effect and could reduce apoptotic cell death after ischemic injury.
Collapse
Affiliation(s)
- Eunjae Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun 58128, Republic of Korea; (E.J.); (H.Y.); (J.H.); (J.C.)
- Jeonnam Bioindustry Foundation Biopharmaceutical Research Center, Hwasun-gun 58141, Republic of Korea
| | - Hee Yu
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun 58128, Republic of Korea; (E.J.); (H.Y.); (J.H.); (J.C.)
- Jeonnam Bioindustry Foundation Biopharmaceutical Research Center, Hwasun-gun 58141, Republic of Korea
| | - Eungpil Kim
- Infrastructure Project Organization for Global Industrialization of Vaccine, Sejong-si 30121, Republic of Korea;
| | - Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun 58128, Republic of Korea; (E.J.); (H.Y.); (J.H.); (J.C.)
| | - Jin Yoo
- Department of Physical Education, Chonnam National University, Gwangju 61186, Republic of Korea;
| | - Jiyun Choi
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun 58128, Republic of Korea; (E.J.); (H.Y.); (J.H.); (J.C.)
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun 58128, Republic of Korea; (E.J.); (H.Y.); (J.H.); (J.C.)
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun 58128, Republic of Korea; (E.J.); (H.Y.); (J.H.); (J.C.)
| |
Collapse
|
3
|
Zhang W, Wang J, Shan C. The eEF1A protein in cancer: Clinical significance, oncogenic mechanisms, and targeted therapeutic strategies. Pharmacol Res 2024; 204:107195. [PMID: 38677532 DOI: 10.1016/j.phrs.2024.107195] [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: 02/28/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Eukaryotic elongation factor 1A (eEF1A) is among the most abundant proteins in eukaryotic cells. Evolutionarily conserved across species, eEF1A is in charge of translation elongation for protein biosynthesis as well as a plethora of non-translational moonlighting functions for cellular homeostasis. In malignant cells, however, eEF1A becomes a pleiotropic driver of cancer progression via a broad diversity of pathways, which are not limited to hyperactive translational output. In the past decades, mounting studies have demonstrated the causal link between eEF1A and carcinogenesis, gaining deeper insights into its multifaceted mechanisms and corroborating its value as a prognostic marker in various cancers. On the other hand, an increasing number of natural and synthetic compounds were discovered as anticancer eEF1A-targeting inhibitors. Among them, plitidepsin was approved for the treatment of multiple myeloma whereas metarrestin was currently under clinical development. Despite significant achievements in these two interrelated fields, hitherto there lacks a systematic examination of the eEF1A protein in the context of cancer research. Therefore, the present work aims to delineate its clinical implications, molecular oncogenic mechanisms, and targeted therapeutic strategies as reflected in the ever expanding body of literature, so as to deepen mechanistic understanding of eEF1A-involved tumorigenesis and inspire the development of eEF1A-targeted chemotherapeutics and biologics.
Collapse
Affiliation(s)
- Weicheng Zhang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China.
| | - Jiyan Wang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China
| | - Changliang Shan
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China.
| |
Collapse
|
4
|
Sarkar A, Singh MP. A Complex Interplay of DJ-1, LRRK2, and Nrf2 in the Regulation of Mitochondrial Function in Cypermethrin-Induced Parkinsonism. Mol Neurobiol 2024; 61:953-970. [PMID: 37674036 DOI: 10.1007/s12035-023-03591-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 09/08/2023]
Abstract
Cypermethrin impairs mitochondrial function, induces redox imbalance, and leads to Parkinsonism in experimental animals. Knockdown of deglycase-1 (DJ-1) gene, which encodes a redox-sensitive antioxidant protein, aggravates cypermethrin-mediated α-synuclein overexpression and oxidative alteration of proteins. DJ-1 is also reported to be essential for maintaining stability of nuclear factor erythroid 2-related factor 2 (Nrf2), shielding cells against oxidative insult. Leucine-rich repeat kinase 2 (LRRK2), another protein associated with Parkinson's disease, is also involved in regulating mitochondrial function. However, underlying molecular mechanisms remain elusive. The study intended to explore an interaction of DJ-1, LRRK2, and Nrf2 in the regulation of mitochondrial function in cypermethrin-induced Parkinsonism. Small interfering RNA-mediated knockdown of DJ-1 and LRRK2 gene and pharmacological activation of Nrf2 were performed in rats and/or human neuroblastoma cells with or without cypermethrin. Indexes of oxidative stress, mitochondrial impairment, and Parkinsonism along with α-synuclein expression, post-translational modification, and aggregation were measured. DJ-1 gene knockdown exacerbated cypermethrin-induced increase in oxidative stress and intrinsic apoptosis and reduction in expression of mitochondrial antioxidant proteins via inhibiting nuclear translocation of Nrf2. Additionally, cypermethrin-induced oxidative stress, mitochondrial impairment, and α-synuclein expression and aggregation were found to be suppressed by LRRK2 gene knockdown, by promoting Nrf2 nuclear translocation and expression of mitochondrial antioxidant proteins. Furthermore, Nrf2 activator, sulforaphane, ameliorated cypermethrin-induced mitochondrial impairment and oxidative stress and provided protection against dopaminergic neuronal death. The findings indicate that DJ-1 and LRRK2 independently alter Nrf2-mediated changes and a complex interplay among DJ-1, LRRK2, and Nrf2 exists in the regulation of mitochondrial function in cypermethrin-induced Parkinsonism.
Collapse
Affiliation(s)
- Alika Sarkar
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Mahendra Pratap Singh
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| |
Collapse
|
5
|
Zhang Y, Wu K, Liu Y, Sun S, Shao Y, Li Q, Sui X, Duan C. UHRF2 promotes the malignancy of hepatocellular carcinoma by PARP1 mediated autophagy. Cell Signal 2023:110782. [PMID: 37356603 DOI: 10.1016/j.cellsig.2023.110782] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/07/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
Autophagy have critical implications in the proliferation and metastasis of HCC. In the current study, we aimed to explore the underlying mechanisms of UHRF2 regulates HCC cell autophagy and HCC progression. We initially determined the relationship between UHRF2 and HCC autophagy, oncogenicity and patient survival through GSEA database and TCGA database. We mainly investigated the effect of UHRF2 on HCC development and autophagy through western blot, electron microscopy, and immunofluorescence. Functionally, UHRF2 was positively involved in the autophagy activation. Overexpression of UHRF2 reduced apoptosis in HCC cells, and promote the malignancy phenotype of HCC both in vitro and in vivo. Mechanistically, PRDX1 bound to UHRF2 and upregulated its protein expression to facilitate the biological function of UHRF2 in HCC. Meanwhile, UHRF2 bound to autophagy-related protein PARP1 and upregulated PARP1 protein level. The results showed that UHRF2 promoted autophagy and contributed to the malignant phenotype of hepatocellular carcinoma by regulating PARP1 levels. In summary, a novel interaction between PRDX1, UHRF2, and PARP1 was revealed, suggesting that UHRF2 could inspire a potential biomarker and potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Yiqi Zhang
- Department of Cell Biology and Genetics, Center for Molecular Medicine and Oncology Research, Chongqing Medical University, Chongqing 400016, China.
| | - Kejia Wu
- Department of Cell Biology and Genetics, Center for Molecular Medicine and Oncology Research, Chongqing Medical University, Chongqing 400016, China.
| | - Yuxin Liu
- Department of Cell Biology and Genetics, Center for Molecular Medicine and Oncology Research, Chongqing Medical University, Chongqing 400016, China.
| | - Shuangling Sun
- Department of Biochemistry, Chongqing Medical and Pharmaceutical College, Chongqing 400016, China
| | - Yue Shao
- Department of Thoracic surgery, the First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Qingxiu Li
- Department of Cell Biology and Genetics, Center for Molecular Medicine and Oncology Research, Chongqing Medical University, Chongqing 400016, China.
| | - Xinying Sui
- Department of Cell Biology and Genetics, Center for Molecular Medicine and Oncology Research, Chongqing Medical University, Chongqing 400016, China.
| | - Changzhu Duan
- Department of Cell Biology and Genetics, Center for Molecular Medicine and Oncology Research, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
6
|
Khwanraj K, Prommahom A, Dharmasaroja P. eEF1A2 siRNA Suppresses MPP+-Induced Activation of Akt and mTOR and Potentiates Caspase-3 Activation in a Parkinson’s Disease Model. ScientificWorldJournal 2023; 2023:1335201. [PMID: 37051183 PMCID: PMC10085650 DOI: 10.1155/2023/1335201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
The tissue-specific protein eEF1A2 has been linked to the development of neurological disorders. The role of eEF1A2 in the pathogenesis of Parkinson’s disease (PD) has yet to be investigated. The aim of this study was to determine the potential neuroprotective effects of eEF1A2 in an MPP+ model of PD. Differentiated SH-SY5Y cells were transfected with eEF1A2 siRNA, followed by MPP+ exposure. The expression of p-Akt1 and p-mTORC1 was determined using Western blotting. The expression of p53, Bax, Bcl-2, and caspase-3 was evaluated using qRT-PCR. Cleaved caspase-3 levels and Annexin V/propidium iodide flow cytometry were used to determine apoptosis. The effects of PI3K inhibition were examined. The results showed that eEF1A2 siRNA significantly reduced the eEF1A2 expression induced by MPP+. MPP+ treatment activated Akt1 and mTORC1; however, eEF1A2 knockdown suppressed this activation. In eEF1A2-knockdown cells, MPP+ treatment increased the expression of p53 and caspase-3 mRNA levels as well as increased apoptotic cell death when compared to MPP+ treatment alone. In cells exposed to MPP+, upstream inhibition of the Akt/mTOR pathway, by either LY294002 or wortmannin, inhibited the phosphorylation of Akt1 and mTORC1. Both PI3K inhibitors increased eEF1A2 expression in cells, whether or not they were also treated with MPP+. In conclusion, eEF1A2 may function as a neuroprotective factor against MPP+, in part by regulating the Akt/mTOR pathway upstream.
Collapse
Affiliation(s)
- Kawinthra Khwanraj
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Athinan Prommahom
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | | |
Collapse
|
7
|
Zhang ZJ, Sun ZX, Liu HJ. EEF1A2 accelerates the protein translation of chemokine in rat myocardial cells induced by ischemia-reperfusion. Heliyon 2023; 9:e15305. [PMID: 37101626 PMCID: PMC10123182 DOI: 10.1016/j.heliyon.2023.e15305] [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: 02/20/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/28/2023] Open
Abstract
How to reduce the damage caused by myocardial ischemia-reperfusion (IR) in a timely manner to save patients' lives is still a great clinical challenge. Although dexmedetomidine (DEX) has been reported to protect the myocardium, the regulatory mechanism of gene translation responding to IR injury and DEX protection is poorly understood. In this study, IR rat model with DEX and the antagonist yohimbine (YOH) pretreatment were established, and RNA sequencing was carried out to seek the important regulators in differential expressed genes. A series of cytokines and chemokine as well as eukaryotic translation elongation factor 1 alpha 2 (EEF1A2) were induced by IR compared to control and compromised by DEX pretreatment compared to IR, then reversed by YOH. Immunoprecipitation was conducted to identify that peroxiredoxin 1 (PRDX1) interacted with EEF1A2 and contributed to the recruitment of EEF1A2 on mRNA molecules of cytokines and chemokine. Knockdown of PRDX1 could weaken the enhancive effect of EEF1A2 for gene translation of IL6, CXCL2 and CXCL11 under the IR condition, and indeed reduce cell apoptosis of cardiomyocytes. We also determined that the RNA motif "USCAGDCU" at 5' UTR could be particularly recognized by PRDX1. Destruction of this motif at the 5' UTR of IL6, CXCL2 and CXCL11 by CRISPR-CAS9 could result in the loss occupancies of EEF1A2 and PRDX1 on the mRNA of these three genes. Our observations showed the importance of PRDX1 in the reasonable control of cytokine and chemokine expression to prevent excessive inflammatory response to cell damage.
Collapse
Affiliation(s)
| | | | - Hai-jian Liu
- Corresponding author. 1500 Zhouyuan Road, Shanghai, 201318, China
| |
Collapse
|
8
|
Gao G, Duan Y, Chang F, Zhang T, Huang X, Yu C. METTL14 promotes apoptosis of spinal cord neurons by inducing EEF1A2 m6A methylation in spinal cord injury. Cell Death Discov 2022; 8:15. [PMID: 35013140 PMCID: PMC8748977 DOI: 10.1038/s41420-021-00808-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/12/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating traumatic condition. METTL14-mediated m6A modification is associated with SCI. This study was intended to investigate the functional mechanism of RNA methyltransferase METTL14 in spinal cord neuron apoptosis during SCI. The SCI rat model was established, followed by evaluation of pathological conditions, apoptosis, and viability of spinal cord neurons. The neuronal function of primary cultured spinal motoneurons of rats was assessed after hypoxia/reoxygenation treatment. Expressions of EEF1A2, Akt/mTOR pathway-related proteins, inflammatory cytokines, and apoptosis-related proteins were detected. EEF1A2 was weakly expressed and Akt/mTOR pathway was inhibited in SCI rat models. Hypoxia/Reoxygenation decreased the viability of spinal cord neurons, promoted LDH release and neuronal apoptosis. EEF1A2 overexpression promoted the viability of spinal cord neurons, inhibited neuronal apoptosis, and decreased inflammatory cytokine levels. Silencing METTL14 inhibited m6A modification of EEF1A2 and increased EEF1A2 expression while METTL14 overexpression showed reverse results. EEF1A2 overexpression promoted viability and inhibited apoptosis of spinal cord neurons and inflammation by activating the Akt/mTOR pathway. In conclusion, silencing METTL14 repressed apoptosis of spinal cord neurons and attenuated SCI by inhibiting m6A modification of EEF1A2 and activating the Akt/mTOR pathway.
Collapse
Affiliation(s)
- Gang Gao
- Department of spinal minimally invasive surgery, Shanxi Provincial People's Hospital, No.29 Shuangtasi Street, Taiyuan City, Shanxi Province, 030012, China.
| | - Yufen Duan
- Department of endocrinology, Shanxi coal central hospital, No.101 Xuefu street, Xiaodian District, Taiyuan City, Shanxi Province, China
| | - Feng Chang
- Department of spinal minimally invasive surgery, Shanxi Provincial People's Hospital, No.29 Shuangtasi Street, Taiyuan City, Shanxi Province, 030012, China
| | - Ting Zhang
- Department of spinal minimally invasive surgery, Shanxi Provincial People's Hospital, No.29 Shuangtasi Street, Taiyuan City, Shanxi Province, 030012, China
| | - Xinhu Huang
- Department of spinal minimally invasive surgery, Shanxi Provincial People's Hospital, No.29 Shuangtasi Street, Taiyuan City, Shanxi Province, 030012, China
| | - Chen Yu
- Department of spinal minimally invasive surgery, Shanxi Provincial People's Hospital, No.29 Shuangtasi Street, Taiyuan City, Shanxi Province, 030012, China
| |
Collapse
|
9
|
Caldi Gomes L, Galhoz A, Jain G, Roser A, Maass F, Carboni E, Barski E, Lenz C, Lohmann K, Klein C, Bähr M, Fischer A, Menden MP, Lingor P. Multi-omic landscaping of human midbrains identifies disease-relevant molecular targets and pathways in advanced-stage Parkinson's disease. Clin Transl Med 2022; 12:e692. [PMID: 35090094 PMCID: PMC8797064 DOI: 10.1002/ctm2.692] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/07/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disorder whose prevalence is rapidly increasing worldwide. The molecular mechanisms underpinning the pathophysiology of sporadic PD remain incompletely understood. Therefore, causative therapies are still elusive. To obtain a more integrative view of disease-mediated alterations, we investigated the molecular landscape of PD in human post-mortem midbrains, a region that is highly affected during the disease process. METHODS Tissue from 19 PD patients and 12 controls were obtained from the Parkinson's UK Brain Bank and subjected to multi-omic analyses: small and total RNA sequencing was performed on an Illumina's HiSeq4000, while proteomics experiments were performed in a hybrid triple quadrupole-time of flight mass spectrometer (TripleTOF5600+) following quantitative sequential window acquisition of all theoretical mass spectra. Differential expression analyses were performed with customized frameworks based on DESeq2 (for RNA sequencing) and with Perseus v.1.5.6.0 (for proteomics). Custom pipelines in R were used for integrative studies. RESULTS Our analyses revealed multiple deregulated molecular targets linked to known disease mechanisms in PD as well as to novel processes. We have identified and experimentally validated (quantitative real-time polymerase chain reaction/western blotting) several PD-deregulated molecular candidates, including miR-539-3p, miR-376a-5p, miR-218-5p and miR-369-3p, the valid miRNA-mRNA interacting pairs miR-218-5p/RAB6C and miR-369-3p/GTF2H3, as well as multiple proteins, such as CHI3L1, HSPA1B, FNIP2 and TH. Vertical integration of multi-omic analyses allowed validating disease-mediated alterations across different molecular layers. Next to the identification of individual molecular targets in all explored omics layers, functional annotation of differentially expressed molecules showed an enrichment of pathways related to neuroinflammation, mitochondrial dysfunction and defects in synaptic function. CONCLUSIONS This comprehensive assessment of PD-affected and control human midbrains revealed multiple molecular targets and networks that are relevant to the disease mechanism of advanced PD. The integrative analyses of multiple omics layers underscore the importance of neuroinflammation, immune response activation, mitochondrial and synaptic dysfunction as putative therapeutic targets for advanced PD.
Collapse
Affiliation(s)
- Lucas Caldi Gomes
- Department of NeurologyRechts der Isar HospitalTechnical University of MunichMünchenGermany
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
| | - Ana Galhoz
- Helmholtz Zentrum München GmbH ‐ German Research Center for Environmental HealthInstitute of Computational BiologyNeuherbergGermany
- Department of BiologyLudwig‐Maximilians University MunichMartinsriedGermany
| | - Gaurav Jain
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - Anna‐Elisa Roser
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
| | - Fabian Maass
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
| | - Eleonora Carboni
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
| | - Elisabeth Barski
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
| | - Christof Lenz
- Institute of Clinical ChemistryUniversity Medical Center GöttingenGöttingenGermany
- Bioanalytical Mass Spectrometry GroupMax Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Katja Lohmann
- Institute of NeurogeneticsUniversity of LübeckLübeckGermany
| | | | - Mathias Bähr
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - André Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
- Department of Psychiatry and PsychotherapyUniversity Medical Center GöttingenGöttingenGermany
| | - Michael P. Menden
- Helmholtz Zentrum München GmbH ‐ German Research Center for Environmental HealthInstitute of Computational BiologyNeuherbergGermany
- Department of BiologyLudwig‐Maximilians University MunichMartinsriedGermany
- German Centre for Diabetes Research (DZD e.V.)NeuherbergGermany
| | - Paul Lingor
- Department of NeurologyRechts der Isar HospitalTechnical University of MunichMünchenGermany
- German Center for Neurodegenerative Diseases (DZNE)MünchenGermany
| |
Collapse
|
10
|
Hu Y, Zhang X, Lian F, Yang J, Xu X. Combination of Lutein and DHA Alleviate H 2O 2 Induced Cytotoxicity in PC12 Cells by Regulating the MAPK Pathway. J Nutr Sci Vitaminol (Tokyo) 2021; 67:234-242. [PMID: 34470998 DOI: 10.3177/jnsv.67.234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Docosahexaenoic acid (DHA) and lutein are important nutrients for brain health. Whether there were synergistic effects of DHA and lutein on the protection against neuronal cell damage induced by oxidative stress remained unclear. The present study was designed to investigate the synergistic effects of DHA and lutein against hydrogen peroxide (H2O2)-induced oxidative challenge in PC12 cells. PC12 cells were divided into different groups and received H2O2 (80 μM), lutein (20 μM)+H2O2 (80 μM), DHA (25 μM)+H2O2 (80 μM), and lutein (20 μM)+DHA (25 μM)+H2O2 (80 μM), respectively. The results indicated that pre-treatment of cells with lutein, DHA and DHA+lutein could significantly antagonize the H2O2-mediated growth inhibition and morphological changes in PC12 cells (p<0.05). Molecularlevel studies indicated that the DHA+lutein combination can significantly inhibit the mRNA expression of AMAD10 and BAX. Furthermore, Western blot analysis demonstrated that DHA+lutein synergistically inhibits the phosphorylation of JNK1/2. The results of the present study suggest that DHA and lutein in combination may be utilized as potent antioxidative compounds, with potential preventative or palliative effects on age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yan Hu
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University
| | - Xu Zhang
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University
| | - Fuzhi Lian
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University
| | - Jun Yang
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University
| | - Xianrong Xu
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University
| |
Collapse
|
11
|
Szeliga M. Peroxiredoxins in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:E1203. [PMID: 33265993 PMCID: PMC7761365 DOI: 10.3390/antiox9121203] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022] Open
Abstract
Substantial evidence indicates that oxidative/nitrosative stress contributes to the neurodegenerative diseases. Peroxiredoxins (PRDXs) are one of the enzymatic antioxidant mechanisms neutralizing reactive oxygen/nitrogen species. Since mammalian PRDXs were identified 30 years ago, their significance was long overshadowed by the other well-studied ROS/RNS defense systems. An increasing number of studies suggests that these enzymes may be involved in the neurodegenerative process. This article reviews the current knowledge on the expression and putative roles of PRDXs in neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and dementia with Lewy bodies, multiple sclerosis, amyotrophic lateral sclerosis and Huntington's disease.
Collapse
Affiliation(s)
- Monika Szeliga
- Mossakowski Medical Research Centre, Department of Neurotoxicology, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| |
Collapse
|