1
|
Wadhwa R, Hegde M, Zhang H, Kaul A, Wang J, Ishida Y, Terao K, Kunnumakkara AB, Kaul SC. Antistress and Antiaging Potentials of Alpha-Lipoic Acid: Insights from Cell Culture-Based Experiments. Appl Biochem Biotechnol 2024; 196:8791-8808. [PMID: 38941028 DOI: 10.1007/s12010-024-04994-4] [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] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
Chronic stress has been linked to a large number of pathologies, including cancer, premature aging, and neurodegenerative diseases. The accumulation of molecular waste resulting from oxidative and heavy metal-induced stress has been ascribed as a major factor contributing to these diseases. With this in mind, we started by screening 13 small molecules to determine their antistress potential in heavy metal stress-exposed C6 glioblastoma and found that alpha-lipoic acid (ALA) (a natural antioxidant abundantly present in yeast, spinach, broccoli, and meat) was the most effective candidate. We then conducted molecular analyses to validate its mechanism of action. Dose-dependent toxicity assays of cells treated with two ALA enantiomers, R-ALA and S-ALA, showed that they are nontoxic and can be tolerated at relatively high doses. Cells exposed to heavy metal, heat, and oxidative stress showed better recovery when cultured in R-ALA-/S-ALA-supplemented medium, supported by reduction of reactive oxygen species (ROS), aggregated proteins, and mitochondrial and deoxyribonucleic acid (DNA) damage. Molecular analyses revealed protection against stress-induced apoptosis and induction of autophagy in R-ALA- and S-ALA-treated C6/U2OS cells. Consistent with these findings, normal human fibroblasts showed lifespan extension. Taken together, this study demonstrates that lipoic acid has antiaging and antistress potential and warrants further attention in laboratory and clinical studies.
Collapse
Affiliation(s)
- Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305-8565, Japan
| | - Mangala Hegde
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT), Guwahati, Assam, 781 039, India
| | - Huayue Zhang
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305-8565, Japan
| | - Ashish Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305-8565, Japan
| | - Jia Wang
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305-8565, Japan
| | - Yoshiyuki Ishida
- CycloChem Bio Co., Ltd., 7-4-5 Minatojima-Minamimachi, Chuo-Ku, Kobe, 650 0047, Japan
| | - Keiji Terao
- CycloChem Bio Co., Ltd., 7-4-5 Minatojima-Minamimachi, Chuo-Ku, Kobe, 650 0047, Japan
| | - Ajaikumar B Kunnumakkara
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT), Guwahati, Assam, 781 039, India.
| | - Sunil C Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305-8565, Japan.
| |
Collapse
|
2
|
Lu W, Chu H, Yang C, Li X. Transcription factor EB (TFEB) promotes autophagy in early brain injury after subarachnoid hemorrhage in rats. Neurosurg Rev 2024; 47:741. [PMID: 39375262 DOI: 10.1007/s10143-024-02879-y] [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: 02/05/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 10/09/2024]
Abstract
Subarachnoid hemorrhage (SAH) has high mortality. Early brain injury (EBI) is responsible for unfavorable outcomes for patients with SAH. The protective involvement of autophagy in hemorrhagic stroke has been proposed. The transcription factor EB (TFEB) can increase autophagic flux by promoting autophagosome formation and autophagosome-lysosome fusion, and dysregulation of TFEB activity might induce the development of several diseases. However, the biological functions of TFEB in EBI after SAH remain unknown. We established an animal model of SAH by the modified endovascular perforation method. Expression of TFEB and autophagy required genes was measured by western blotting and immunofluorescence staining. SAH grading, brain water content and neurobehavioral functions were evaluated at 24 h post-SAH. Neuronal apoptosis in cerebral cortex was assessed by TUNEL staining and Fluoro Jade B staining. TFEB was downregulated in SAH rats, and its overexpression reduced brain edema and ameliorated neurological deficits of SAH rats. Additionally, the neuronal apoptosis induced by SAH was inhibited by TFEB overexpression. Moreover, TFEB overexpression promoted autophagy after SAH. TFEB overexpression promotes autophagy to inhibit neuronal apoptosis, brain edema and neurological deficits post-SAH.
Collapse
Affiliation(s)
- Wenqi Lu
- Department of Anesthesiology, The first Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| | - Haichao Chu
- Department of Anesthesiology, The first Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| | - Chunchen Yang
- Department of Anesthesiology, The first Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| | - Xiaoxu Li
- Department of Neurosurgery, The first Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China.
| |
Collapse
|
3
|
Ding PF, Liu XZ, Peng Z, Cui Y, Liu Y, Zhang JT, Zhu Q, Wang J, Zhou Y, Gao YY, Hang CH, Li W. miR-93-5p impairs autophagy-lysosomal pathway via TET3 after subarachnoid hemorrhage. Exp Neurol 2024; 380:114904. [PMID: 39094768 DOI: 10.1016/j.expneurol.2024.114904] [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: 05/07/2024] [Revised: 07/01/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Intact autophagy-lysosomal pathway (ALP) in neuronal survival is crucial. However, it remains unclear whether ALP is intact after subarachnoid hemorrhage (SAH). Ten-eleven translocation (TET) 3 primarily regulates genes related to autophagy in neurons in neurodegenerative diseases. This study aims to investigate the role of TET3 in the ALP following SAH. The results indicate that the ALP is impaired after SAH, with suppressed autophagic flux and an increase in autophagosomes. This is accompanied by a decrease in TET3 expression. Activation of TET3 by α-KG can improve ALP function and neural function to some extent. Silencing TET3 in neurons significantly inhibited the ALP function and increased apoptosis. Inhibition of miR-93-5p, which is elevated after SAH, promotes TET3 expression. This suggests that the downregulation of TET3 after SAH is, at least in part, due to elevated miR-93-5p. This study clarifies the key role of TET3 in the functional impairment of the ALP after SAH. The preliminary exploration revealed that miR-93-5p could lead to the downregulation of TET3, which could be a new target for neuroprotective therapy after SAH.
Collapse
Affiliation(s)
- Peng-Fei Ding
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Xun-Zhi Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Zheng Peng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Yue Cui
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Yang Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Jia-Tong Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Qi Zhu
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Jie Wang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Yan Zhou
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Yong-Yue Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| |
Collapse
|
4
|
Zeng J, Fang Z, Duan J, Zhang Z, Wang Y, Wang Y, Chen L, Wang J, Liu F. Activation of Piezo1 by intracranial hypertension induced neuronal apoptosis via activating hippo pathway. CNS Neurosci Ther 2024; 30:e14872. [PMID: 39328029 PMCID: PMC11427798 DOI: 10.1111/cns.14872] [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: 01/15/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 09/28/2024] Open
Abstract
AIM Most of the subarachnoid hemorrhage (SAH) patients experienced the symptom of severe headache caused by intracranial hypertension. Piezo1 is a mechanosensitive ion channel protein. This study aimed to investigate the effect of Piezo1 on neurons in response to intracranial hypertension. METHODS The SAH rat model was performed by the modified endovascular perforation method. Piezo1 inhibitor GsMTx4 was administered intraperitoneally after SAH induction. To investigate the underlying mechanism, the selective Piezo1 agonist Yoda1, Piezo1 shRNA, and MY-875 were administered via intracerebroventricular injection before SAH induction. In vitro, we designed a pressurizing device to exclusively explore the effect of Piezo1 activation on primary neurons. Neurons were pretreated with Piezo1 inhibition followed by intracranial hypertension treatment, and then apoptosis-related proteins were detected. RESULTS Piezo1 inhibition significantly attenuated neuronal apoptosis and improved the outcome of neurological deficits in rats after SAH. The Hippo pathway agonist MY-875 reversed the anti-apoptotic effects of Piezo1 knockdown. In vitro, intracranial hypertension mimicked by the pressurizing device induced Piezo1 expression, resulting in Hippo pathway activation and neuronal apoptosis. The Hippo pathway inhibitor Xmu-mp-1 attenuated Yoda1-induced neuronal apoptosis. In addition, the combination of hypertension and oxyhemoglobin treatment exacerbated neuronal apoptosis. CONCLUSIONS Intracranial hypertension induced Piezo1 expression, neuronal apoptosis, and the Hippo pathway activation; the Hippo signaling pathway is involved in Piezo1 activation-induced neuronal apoptosis in respond to intracranial hypertension. Primary neurons treated with intracranial hypertension and oxyhemoglobin together can better characterize the circumstance of SAH in vivo, which is contributed to construct an ideal in vitro SAH model.
Collapse
Affiliation(s)
- Jia Zeng
- Department of Neurosurgery, Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Zhen Fang
- Department of Neurosurgery, Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Jiajia Duan
- Department of Neurosurgery, Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Zichen Zhang
- Department of Neurosurgery, Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Yunzhi Wang
- Department of Neurosurgery, Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Yiping Wang
- Department of Neurosurgery, Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Lei Chen
- Department of Neurosurgery, Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Jikai Wang
- Department of Neurosurgery, Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Fei Liu
- Department of Neurosurgery, Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| |
Collapse
|
5
|
Zhang H, Ren K, Hu Y, Liu B, He Y, Xu H, Ma K, Tian W, Dai L, Zhao D. Neuritin promotes autophagic flux by inhibiting the cGAS-STING pathway to alleviate brain injury after subarachnoid haemorrhage. Brain Res 2024; 1836:148909. [PMID: 38570154 DOI: 10.1016/j.brainres.2024.148909] [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: 03/01/2024] [Accepted: 03/31/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Early brain injury (EBI) is closely associated with poor prognosis in patients with subarachnoid haemorrhage (SAH), with autophagy playing a pivotal role in EBI. However, research has shown that the stimulator of interferon genes (STING) pathway impacts autophagic flux. While the regulatory impact of neuritin on EBI and autophagic flux has been established previously, the underlying mechanism remains unclear. This study aimed to determine the role of the cGAS-STING pathway in neuritin-mediated regulation of autophagic flux following SAH. METHODS A SAH model was established in male Sprague-Dawley rats via intravascular perforation. Neuritin overexpressions using adeno-associated virus, the STING antagonist "C-176," and the activator, "CMA," were determined to investigate the cGAS-STING pathway's influence on autophagic flux and brain injury post-SAH, along with the neuritin's regulatory effect on STING. In this study, SAH grade, neurological score, haematoxylin and eosin (H&E) staining, brain water content (BWC), sandwich enzyme-linked immunosorbent assay, Evans blue staining, immunofluorescence staining, western blot analysis, and transmission electron microscopy (TEM) were examined. RESULTS Neuritin overexpression significantly ameliorated neurobehavioural scores, blood-brain barrier injury, brain oedema, and impaired autophagic flux in SAH-induced rats. STING expression remarkably increased post-SAH. C-176 and CMA mitigated and aggravated autophagic flux injury and brain injury, respectively, while inhibiting and enhancing STING, respectively. Particularly, CMA treatment nullified the protective effects of neuritin against autophagic flux and mitigated brain injury. CONCLUSION Neuritin alleviated EBI by restoring impaired autophagic flux after SAH through the regulation of the cGAS-STING pathway.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Shihezi University, Shihezi 832000, China
| | - Kunhao Ren
- Department of Neurosurgery, the First Affiliated Hospital of Shihezi University, Shihezi 832000, China
| | - Youjie Hu
- Department of Neurosurgery, the First Affiliated Hospital of Shihezi University, Shihezi 832000, China
| | - Bin Liu
- Department of Neurosurgery, the First Affiliated Hospital of Shihezi University, Shihezi 832000, China
| | - Yaowen He
- Department of Neurosurgery, the First Affiliated Hospital of Shihezi University, Shihezi 832000, China
| | - Hui Xu
- Department of Neurosurgery, the First Affiliated Hospital of Shihezi University, Shihezi 832000, China
| | - Ketao Ma
- Shihezi University School of Medicine, Shihezi 832000, China
| | - Weidong Tian
- Department of Neurosurgery, the First Affiliated Hospital of Shihezi University, Shihezi 832000, China
| | - Linzhi Dai
- Department of Neurosurgery, the First Affiliated Hospital of Shihezi University, Shihezi 832000, China.
| | - Dong Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Shihezi University, Shihezi 832000, China.
| |
Collapse
|
6
|
Lauzier DC, Jayaraman K, Yuan JY, Diwan D, Vellimana AK, Osbun J, Chatterjee AR, Athiraman U, Dhar R, Zipfel GJ. Early Brain Injury After Subarachnoid Hemorrhage: Incidence and Mechanisms. Stroke 2023; 54:1426-1440. [PMID: 36866673 PMCID: PMC10243167 DOI: 10.1161/strokeaha.122.040072] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Aneurysmal subarachnoid hemorrhage is a devastating condition causing significant morbidity and mortality. While outcomes from subarachnoid hemorrhage have improved in recent years, there continues to be significant interest in identifying therapeutic targets for this disease. In particular, there has been a shift in emphasis toward secondary brain injury that develops in the first 72 hours after subarachnoid hemorrhage. This time period of interest is referred to as the early brain injury period and comprises processes including microcirculatory dysfunction, blood-brain-barrier breakdown, neuroinflammation, cerebral edema, oxidative cascades, and neuronal death. Advances in our understanding of the mechanisms defining the early brain injury period have been accompanied by improved imaging and nonimaging biomarkers for identifying early brain injury, leading to the recognition of an elevated clinical incidence of early brain injury compared with prior estimates. With the frequency, impact, and mechanisms of early brain injury better defined, there is a need to review the literature in this area to guide preclinical and clinical study.
Collapse
Affiliation(s)
- David C. Lauzier
- Department of Neurological Surgery, Washington University School of Medicine
| | - Keshav Jayaraman
- Department of Neurological Surgery, Washington University School of Medicine
| | - Jane Y. Yuan
- Department of Neurological Surgery, Washington University School of Medicine
| | - Deepti Diwan
- Department of Neurological Surgery, Washington University School of Medicine
| | - Ananth K. Vellimana
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
- Mallinckrodt Institute of Radiology, Washington University School of Medicine
| | - Joshua Osbun
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
- Mallinckrodt Institute of Radiology, Washington University School of Medicine
| | - Arindam R. Chatterjee
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
- Mallinckrodt Institute of Radiology, Washington University School of Medicine
| | | | - Rajat Dhar
- Department of Neurology, Washington University School of Medicine
| | - Gregory J. Zipfel
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
| |
Collapse
|
7
|
Yang X, Han M, Wang X, Wang J, Sun X, Zhang C, Yan S, Huang L, Chen Y. Evaluation of the synergistic effects of epigallocatechin-3-gallate-loaded PEGylated-PLGA nanoparticles with nimodipine against neuronal injury after subarachnoid hemorrhage. Front Nutr 2023; 9:953326. [PMID: 36687668 PMCID: PMC9845867 DOI: 10.3389/fnut.2022.953326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 11/25/2022] [Indexed: 01/05/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a devastating subtype of stroke with high mortality and morbidity. Although serious side effects might occur, nimodipine, a second-generation 1,4-dihydropyridine calcium channel blocker, is clinically used to improve neurological outcomes after SAH. Recently, (-)-epigallocatechin-3-gallate (EGCG) has been reported to inhibit Ca2+ overloading-induced mitochondrial dysfunction, oxidative stress, and neuronal cell death after SAH; however, low bioavailability, instability, and cytotoxicity at a high dose limited the clinical application of EGCG. To overcome these limitations, PEGylated-PLGA EGCG nanoparticles (EGCG-NPs) were constructed to enhance the bioavailability by using the double-emulsion method. Antioxidative activity, cytotoxicity, behavioral, and immunohistochemistry studies were carried out to determine the neuroprotective effectiveness after cotreatment with EGCG-NPs (75 mg/kg/d preconditioning for 7 days before SAH) and nimodipine (10 mg/kg/d after 30 min of SAH) by using in vivo SAH models. The optimized EGCG-NPs with a Box-Behnken design showed a small particle size of 167 nm, a zeta potential value of -22.6 mV, an encapsulation efficiency of 86%, and a sustained-release profile up to 8 days in vitro. Furthermore, EGCG-NPs (75 mg/kg/d) had superior antioxidative activity to free EGCG (100 mg/kg/d). EGCG-NPs combined with nimodipine exhibited significant synergistic effects against neuronal cell death by suppressing oxidative stress, Ca2+ overloading, mitochondrial dysfunction, and autophagy after SAH. These results suggest that cotreatment with EGCG-NPs and nimodipine may serve as a promising novel strategy for the treatment of SAH.
Collapse
Affiliation(s)
- Xianguang Yang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Mengguo Han
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Xue Wang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Jian Wang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Xiaoxue Sun
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Chunyan Zhang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Shuaiguo Yan
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Liyong Huang
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Henan, China,Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Henan, China,Liyong Huang ✉
| | - Ying Chen
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China,*Correspondence: Ying Chen ✉
| |
Collapse
|
8
|
Zhao HX, Li X, Liu JL, Guan GQ, Dan XG. Metabolomic profiling of bovine leucocytes transformed by Theileria annulata under BW720c treatment. Parasit Vectors 2022; 15:356. [PMID: 36199104 PMCID: PMC9533618 DOI: 10.1186/s13071-022-05450-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Background When Theileria annulata infects host cells, it undertakes unlimited proliferation as tumor cells. Although the transformed cells will recover their limited reproductive characteristics and enter the apoptosis process after treatment with buparvaquone (BW720c), the metabolites and metabolic pathways involved are not clear. Methods The transformed cells of T. annulata were used as experimental materials, and the buparvaquone treatment group and DMSO control group were used. Qualitative and quantitative analysis was undertaken of 36 cell samples based on the LC–QTOF platform in positive and negative ion modes. The metabolites of the cell samples after 72 h of drug treatment were analyzed, as were the different metabolites and metabolic pathways involved in the BW720c treatment. Finally, the differential metabolites and metabolic pathways in the transformed cells were found. Results A total of 1425 metabolites were detected in the negative ion mode and 1298 metabolites were detected in the positive ion mode. After drug treatment for 24 h, 48 h, and 72 h, there were 56, 162, and 243 differential metabolites in negative ion mode, and 35, 121, and 177 differential metabolites in positive ion mode, respectively. These differential metabolites are mainly concentrated on various essential amino acids. Conclusion BW720c treatment induces metabolic disturbances in T. annulata-infected cells by regulating the metabolism of leucine, arginine, and l-carnitine, and induces host cell apoptosis. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05450-0.
Collapse
Affiliation(s)
- Hong-Xi Zhao
- School of Agriculture, Ningxia University, Yinchuan, 750021, People's Republic of China.
| | - Xia Li
- School of Agriculture, Ningxia University, Yinchuan, 750021, People's Republic of China
| | - Jun-Long Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, 730046, People's Republic of China
| | - Gui-Quan Guan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, 730046, People's Republic of China
| | - Xin-Gang Dan
- School of Agriculture, Ningxia University, Yinchuan, 750021, People's Republic of China.
| |
Collapse
|
9
|
Prediction Model between Serum Vitamin D and Neurological Deficit in Cerebral Infarction Patients Based on Machine Learning. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2914484. [PMID: 35799673 PMCID: PMC9256304 DOI: 10.1155/2022/2914484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/27/2022] [Accepted: 06/15/2022] [Indexed: 11/22/2022]
Abstract
Objective Vitamin D is associated with neurological deficits in patients with cerebral infarction. This study uses machine learning to evaluate the prediction model's efficacy of the correlation between vitamin D and neurological deficit in patients with cerebral infarction. Methods A total of 200 patients with cerebral infarction admitted to the Department of Neurology of our hospital from July 2018 to June 2019 were selected. The patients were randomly divided into a training set (n = 140) and a test set (n = 60) in a 7 : 3 ratio. The prediction model is constructed from the training set's data, and the model's prediction effect was evaluated by test set data. The area under the receiver operator characteristic curve was used to assess the prediction efficiency of models. Results In the training set, the area under the curve (AUC) of the logistic regression model and XGBoost algorithm model was 0.727 (95% CI: 0.601~0.854) and 0.818 (95% CI: 0.734~0.934), respectively. While in the test set, the AUC of the logistic regression model and XGBoost algorithm model was 0.761 (95% CI: 0.640~0.882) and 0.786 (95% CI: 0.670~0.902), respectively. Conclusion The prediction model of the correlation between vitamin D and neurological deficit in patients with cerebral infarction based on machine learning has a good prediction efficiency.
Collapse
|
10
|
Hao H, Bai Y, Liu Y, Liang J, Guo S. Protective mechanism of FoxO1 against early brain injury after subarachnoid hemorrhage by regulating autophagy. Brain Behav 2021; 11:e2376. [PMID: 34661985 PMCID: PMC8613423 DOI: 10.1002/brb3.2376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 09/02/2021] [Accepted: 09/13/2021] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Early brain injury (EBI) plays a key role in the devastating outcomes after subarachnoid hemorrhage (SAH). Autophagy and apoptosis may share a common molecular inducer that regulates the process of cell death. FoxO1, as a key regulator of neuronal autophagy which is involved in apoptosis, has not been reported in SAH rats. This work was to investigate the protective and anti-inflammatory effects of FoxO1 on EBI after SAH by regulating autophagy. METHODS In this study, we constructed the SAH model. In experiment I, low dose (50 μl of 1 × 108 IU/ml) or high dose (50 μl of 1 × 1010 IU/ml) of FoxO1 gene overexpressed adenovirus vector was injected into the lateral ventricle of rats before SAH. In experiment II, we reported the effect of FoxO1 overexpress on nerve function recovery, oedema, BBB leakage, neuronal death in rats after SAH through autophagy regulation. Post-SAH evaluation included neurological function score, brain water content, evans blue exosmosis, pathological changes, inflammatory response and apoptosis. RESULTS The experiment I showed that either low or high dose of ad-FoxO1 could significantly improve nerve function, reduce cerebral water content and reduce blood-brain barrier (BBB) destruction in rats, indicating that ad-FoxO1 had a protective effect on brain injury in rats EBI after SAH. In addition, ad-FoxO1 promoted autophagy in rat hippocampal tissue, as evidenced by accumulation of LC3II/I and Beclin-1 and degradation of p62. Furthermore, ad-FoxO1 inhibited the inflammatory response and apoptosis of rat hippocampal neurons after SAH. The experiment II showed that both ad-FoxO1 and rapamycin attenuated the injury of nerve function in rats after SAH, and this synergistic effect further reduced cerebral edema and evansblue extravasation, decreased hippocampus neuronal cell apoptosis, and declined inflammatory response. However, this was contrary to the results of chloroquine. These findings suggested that FoxO1 regulated the neural function of EBI after SAH through the autophagy pathway. CONCLUSIONS The findings in this study was beneficial for identifying the novel therapeutic target for the treatment of SAH.
Collapse
Affiliation(s)
- Haitao Hao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China.,Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, P. R. China
| | - Yahui Bai
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Yu Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Junxin Liang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Shichao Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| |
Collapse
|