1
|
Gong Y, Wu M, Huang Y, He X, Yuan J, Dang B. Research developments in the neurovascular unit and the blood‑brain barrier (Review). Biomed Rep 2025; 22:88. [PMID: 40166412 PMCID: PMC11956146 DOI: 10.3892/br.2025.1966] [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: 11/18/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025] Open
Abstract
The neurovascular unit (NVU) is composed of neurons, glial cells, brain microvascular endothelial cells (BMECs), pericytes, and the extracellular matrix. The NVU controls the permeability of the blood-brain barrier (BBB) and protects the brain from harmful blood-borne and endogenous and exogenous substances. Among these, neurons transmit signals, astrocytes provide nutrients, microglia regulate inflammation, and BMECs and pericytes strengthen barrier tightness and coverage. These cells, due to their physical structure, anatomical location, or physiological function, maintain the microenvironment required for normal brain function. In this review, the BBB structure and mechanisms are examined to obtain a better understanding of the factors that influence BBB permeability. The findings may aid in safeguarding the BBB and provide potential therapeutic targets for drugs affecting the central nervous system.
Collapse
Affiliation(s)
- Yating Gong
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Muyao Wu
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Yaqian Huang
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Xiaoyi He
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Jiaqi Yuan
- Department of Neurosurgery, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Baoqi Dang
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| |
Collapse
|
2
|
钟 鹏, 胡 晓, 王 振. [Optical coherence tomography angiography and microvessel density quantification in penumbra after traumatic brain injury in rats]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2025; 57:262-266. [PMID: 40219554 PMCID: PMC11992443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Indexed: 04/14/2025]
Abstract
OBJECTIVE To observe the dynamic changes of microvascular injury and repair in the penumbra of traumatic brain injury (TBI) rats with effective cerebral perfusion microvascular imaging using optical coherence tomography angiography (OCTA). METHODS Transparent closed cranial windows were placed in craniotomy rats after TBI caused by weight drop. All the rats in TBI group and control group underwent head MRI examination on the first postoperative day, and the changes of cerebral cortical microvessel density were measured by OCTA through cranial windows on d0, d2, d4, d6, and d8. On the second day after the operation, the same number of rats in the two groups were selected to complete the immunohistochemical staining of brain tissue with pimonidazole, an indicator of hypoxia. RESULTS MRI T2W1 and immunohistochemical staining demonstrated that edema and hypoxia in the traumatic brain tissue extended deeply throughout the entire cortex. OCTA showed that the cortical surface veins of the rats in both groups were significantly dilated and tortuous after operation, and recovered to the postoperative day level on d8. The effective perfusion microvessel density of the rats in both groups gradually recovered after a temporary decrease, and the TBI group decreased from 39.38%±4.48% on d0 to 27.84%±6.01% on d2, which was significantly lower than that on d0, d6, and d8 (P < 0.05). The highest value was 61.71%±7.69% on d8, which was significantly higher than that on d0, d2, and d4 (P < 0.05). The control group decreased from 44.59%±7.78% on d0 to 36.69%±5.49% on d2, which was significantly lower than that on d0, d6, and d8 (P < 0.05). The highest value was 51.92%±5.96% on d8, which was significantly higher than that on d2, and d4 (P < 0.05). Comparing the two groups, the effective perfusion microvessel density in the TBI group was significantly lower than that in the control group on d2 (P=0.021), and significantly higher than that in the control group on d8 (P=0.030). CONCLUSION OCTA can be used as a method of imaging and measurement of effective perfusion microvessels in the injured cerebral cortex of TBI rats. After TBI, the effective perfusion microvessel density in the wound penumbra gradually recovered after decreasing, and increased significantly on d8.
Collapse
Affiliation(s)
- 鹏 钟
- 首都医科大学附属北京友谊医院超声医学科,北京 100050Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - 晓丹 胡
- 北京大学基础医学院,北京 100191School of Basic Medical Sciences, Peking University, Beijing 100191, China
- 北京大学人民医院创伤救治中心,创伤救治与神经再生教育部重点实验室(北京大学),国家创伤医学中心 100044Trauma Center, National Center for Trauma Medicine, Key Laboratory of Trauma and Neural Regeneration, Peking University People's Hospital, Beijing, 100044, China
| | - 振洲 王
- 北京大学人民医院创伤救治中心,创伤救治与神经再生教育部重点实验室(北京大学),国家创伤医学中心 100044Trauma Center, National Center for Trauma Medicine, Key Laboratory of Trauma and Neural Regeneration, Peking University People's Hospital, Beijing, 100044, China
| |
Collapse
|
3
|
Mei Y, She F, Zhang L, Kim G, Li R, Zheng X, Wang Z, Chen R, Wang L, Chen D, Kim J, Zhang T, Lee TH. Zipper-interacting protein kinase mediates neuronal cell death and cognitive dysfunction in traumatic brain injury via regulating DEDD. Cell Death Dis 2025; 16:151. [PMID: 40032841 DOI: 10.1038/s41419-025-07474-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/07/2025] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
Neuronal cell death is a causative process in traumatic brain injury (TBI)-induced structural and functional impairment of the central nervous system. However, the upstream trigger of TBI-induced neuronal loss and the underlying molecular pathways remain unclear. Zipper-interacting protein kinase (ZIPK) has been shown to be upregulated in Alzheimer's disease and ischemic stroke and to play a role in cellular apoptosis, while its pathological significance in TBI has not been reported. Herein, we discovered for the first time that ZIPK expression was markedly elevated in neurons after TBI and that ZIPK caused massive neuronal apoptosis in peri-contusional brain regions. Zipk haploinsufficiency antagonized neuronal cell death and reversed several typical neuropathological changes induced by TBI. Mechanistically, we found that ZIPK affected neuronal viability by modulating death effector domain-containing DNA binding protein (DEDD) and caspase-3 pathway. Specifically, ZIPK could bind to and phosphorylate DEDD at the S9 residue, thus enhancing the stability of DEDD, and leading to the activation of caspase-3-mediated apoptotic cascade in neurons. The rescue of neuronal loss by ZIPK downregulation effectively alleviated TBI-induced behavioral deficits by preserving motor and cognitive abilities in vivo, supporting the decisive role of ZIPK dysregulation in TBI-associated neuronal dysfunctions by modulating neuronal survival. Furthermore, pharmacological suppression of ZIPK activity by a specific inhibitor prior to TBI protected neurons from brain injury-induced cell death and neuronal degeneration in vitro and in vivo by preventing DEDD upregulation and caspase-3 activation. In conclusion, our data reveal the essential contribution of ZIPK to TBI-induced neuronal cell death through the DEDD/caspase-3 cascade, and suggest the potential of targeting ZIPK as an effective strategy for treating TBI-related neuropathologies.
Collapse
Affiliation(s)
- Yingxue Mei
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Fei She
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ling Zhang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Gamin Kim
- Laboratory of Molecular and Cellular Biology, Department of Life Science, Sogang University, Seoul, Korea
| | - Ruomeng Li
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xiuzhi Zheng
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Zonghai Wang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Renxuan Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Long Wang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Dongmei Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jungho Kim
- Laboratory of Molecular and Cellular Biology, Department of Life Science, Sogang University, Seoul, Korea
| | - Tao Zhang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Tae Ho Lee
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
4
|
Wang Y, Fang J, Yuan Q, Yu J, Hu J. GPX3 as a Novel and Potential Therapeutic Target in the Shared Molecular Mechanisms of Traumatic Brain Injury and Parkinson's Disease. J Inflamm Res 2025; 18:1911-1928. [PMID: 39935526 PMCID: PMC11812561 DOI: 10.2147/jir.s506891] [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: 11/17/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
Background Traumatic brain injury (TBI) is a prevalent neurological disorder associated with significant public health burdens and long-term risks, including neurodegenerative diseases such as Parkinson's disease (PD). Emerging evidence suggests a strong link between moderate to severe TBI and an elevated risk of PD, though the underlying mechanisms remain poorly understood. Materials and Methods Common differentially expressed genes (DEGs) were identified in GEO datasets of patients with traumatic brain injury (TBI) and Parkinson's disease (PD). Further analyses, including GO and KEGG pathway enrichment, protein-protein interaction (PPI) network construction, hub gene identification, as well as miRNA and transcription factor prediction and drug candidate screening, were conducted. Subsequently, the expression of hub genes was validated using additional TBI- and PD-related GEO datasets and the Comparative Toxicogenomics Database (CTD). Finally, the expression of hub genes was further validated in a mouse model of TBI induced by controlled cortical impact (CCI). Results Shared transcriptional signatures between TBI and PD were uncovered, highlighting overlapping molecular networks and pathways. The glutathione peroxidase 3 (GPX3) gene emerged as a pivotal hub gene, with its expression significantly altered in both TBI and PD datasets. Conclusion This study underscores the critical role of GPX3 in the molecular intersection of TBI and PD, suggesting it as a novel and potential therapeutic target, offering new insights into potential therapeutic strategies.
Collapse
Affiliation(s)
- Yue Wang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Neurosurgical Institute of Fudan University, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Clinical Medical Center of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
| | - Jiang Fang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Neurosurgical Institute of Fudan University, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Clinical Medical Center of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
| | - Qiang Yuan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Neurosurgical Institute of Fudan University, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Clinical Medical Center of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
| | - Jian Yu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Neurosurgical Institute of Fudan University, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Clinical Medical Center of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
| | - Jin Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Neurosurgical Institute of Fudan University, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
- Shanghai Clinical Medical Center of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
| |
Collapse
|
5
|
Garcia JP, Armbruster M, Sommer M, Nunez-Beringer A, Dulla CG. Glutamate uptake is transiently compromised in the perilesional cortex following controlled cortical impact. Cereb Cortex 2025; 35:bhaf031. [PMID: 40007051 DOI: 10.1093/cercor/bhaf031] [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: 08/28/2024] [Revised: 12/03/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Glutamate, the primary excitatory neurotransmitter in the central nervous system (CNS), is regulated by the excitatory amino acid transporters glutamate transporter 1 (GLT-1) and glutamate aspartate transporter (GLAST). Following traumatic brain injury, extracellular glutamate levels increase, contributing to excitotoxicity, circuit dysfunction, and morbidity. Increased neuronal glutamate release and compromised astrocyte-mediated uptake contribute to elevated glutamate, but the mechanistic and spatiotemporal underpinnings of these changes are not well established. Using the controlled cortical impact model of TBI and iGluSnFR glutamate imaging, we quantified extracellular glutamate dynamics after injury. Three days postinjury, glutamate release was increased, and glutamate uptake and GLT-1 expression were reduced. Seven and 14 days postinjury, glutamate dynamics were comparable between sham and controlled cortical impact animals. Changes in peak glutamate response were unique to specific cortical layers and proximity to injury. This was likely driven by increases in glutamate release, which was spatially heterogeneous, rather than reduced uptake, which was spatially uniform. The astrocyte K+ channel, Kir4.1, regulates activity-dependent slowing of glutamate uptake. Surprisingly, Kir4.1 was unchanged after controlled cortical impact and accordingly, activity-dependent slowing of glutamate uptake was unaltered. This dynamic glutamate dysregulation after traumatic brain injury underscores a brief period in which disrupted glutamate uptake may contribute to dysfunction and highlights a potential therapeutic window to restore glutamate homeostasis.
Collapse
Affiliation(s)
- Jacqueline P Garcia
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
- Cellular, Molecular, and Developmental Biology Program, Tufts University School of Medicine, Boston, MA, United States
| | - Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Mary Sommer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Aliana Nunez-Beringer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
6
|
Bi Y, Luo L, Duan P, Jin Z, Zhang X, He G, Li X, Feng W, Zhang B. Tetrahydrocurcumin exhibits neuroprotective effects by inhibiting neuron ferroptosis via activity of iPLA2β/p38 MAPK phosphorylation in rat TBI model. Free Radic Res 2025; 59:152-168. [PMID: 39936602 DOI: 10.1080/10715762.2025.2465282] [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: 07/03/2024] [Revised: 01/10/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Ferroptosis characterized by iron-dependent lipid peroxidation induced by traumatic brain injury (TBI) is an important factor that aggravates diseases. Studies have shown that tetrahydrocurcumin (THC) has neuroprotective effects in brain injury. However, whether THC inhibits neurocyte ferroptosis after TBI and its mechanism remains unclear. To investigate this, a weight-drop model in rats and H2O2 induced oxidative stress model in SH-SY5Y cells were established, and THC was used for treatment. Immunohistochemical staining showed that iron deposition reached its peak at 8th day after TBI. We found that THC remarkably inhibited iron accumulation in the cortical cortex and corpus callosum, improved neurological damage, reduced acute cerebral edema, weight loss, oxidative stress, and inflammation. Furthermore, the activity of iPLA2β was significantly reduced, and phosphorylation of p38 was increased after TBI, while THC alleviated the decrease in iPLA2β activity and increase in the level of P-p38. It confirmed that THC effectively mitigated ferroptosis, while iPLA2β inhibitor s-BEL could reverse the effects of THC on ferroptosis in vivo and in vitro experiments. In addition, SB202190 which is an inhibitor of p38 could enhance THC protection and lessen formation of ferroptosis-related proteins in cells. In conclusion, these findings suggested that THC may promote neurological function recovery after TBI by inhibiting neuron ferroptosis via activity of iPLA2β/P-p38.
Collapse
Affiliation(s)
- Yonghong Bi
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China
| | - Lan Luo
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengyu Duan
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhehao Jin
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoqian Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guanghui He
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyan Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weiyu Feng
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China
| |
Collapse
|
7
|
Liu L, Jia P, Liu T, Liang J, Dang Y, Rastegar-Kashkooli Y, Li Q, Liu J, Man J, Zhao T, Xing N, Wang F, Chen X, Zhang J, Jiang C, Zille M, Zhang Z, Fan X, Wang J, Wang J. Metabolic dysfunction contributes to mood disorders after traumatic brain injury. Ageing Res Rev 2025; 104:102652. [PMID: 39746403 DOI: 10.1016/j.arr.2024.102652] [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: 09/05/2024] [Revised: 12/15/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Traumatic brain injury (TBI) presents significant risks concerning mortality and morbidity. Individuals who suffer from TBI may exhibit mood disorders, including anxiety and depression. Both preclinical and clinical research have established correlations between TBI and disturbances in the metabolism of amino acids, lipids, iron, zinc, and copper, which are implicated in the emergence of mood disorders post-TBI. The purpose of this review is to elucidate the impact of metabolic dysfunction on mood disorders following TBI and to explore potential strategies for mitigating anxiety and depression symptoms. We researched the PubMed and Web of Science databases to delineate the mechanisms by which metabolic dysfunction contributes to mood disorders in the context of TBI. Particular emphasis was placed on the roles of glutamate, kynurenine, lipids, iron, zinc, and copper metabolism. Metabolic dysfunction is linked to mood disorders post-TBI through multiple pathways, encompassing the glutamatergic system, the kynurenine pathway, endocannabinoids, iron deposition, iron-related ferroptosis, zinc deficiency, and copper dysregulation. Furthermore, this review addresses the influence of metabolic dysfunction on mood disorders in the elderly demographic following TBI. Targeting metabolic dysfunction for therapeutic intervention appears promising in alleviating symptoms of anxiety and depression that arise after TBI. While further investigation is warranted to delineate the underlying pathophysiologic mechanisms of mood disorders post-TBI, current evidence underscores the potential contribution of metabolic dysfunction to these conditions. Therefore, rectifying metabolic dysfunction represents a viable and strategic approach to addressing mood disorders following TBI.
Collapse
Affiliation(s)
- Lang Liu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Peijun Jia
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Tongzhou Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiaxin Liang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Yijia Dang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Qiang Li
- Department of Neurology, Shanghai Gongli Hospital of Pudong New Area, Shanghai 200135, China.
| | - Jingqi Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiang Man
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Ting Zhao
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Fushun Wang
- Department of Psychology, Sichuan Normal University, Chengdu, Sichuan 610060, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiewen Zhang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Chao Jiang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna 1090, Austria.
| | - Zhenhua Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
8
|
Shi H, Song L, Wu Y, Shen R, Zhang C, Liao X, Wang Q, Zhu J. Edaravone Alleviates Traumatic Brain Injury by Inhibition of Ferroptosis via FSP1 Pathway. Mol Neurobiol 2024; 61:10448-10461. [PMID: 38733490 PMCID: PMC11584507 DOI: 10.1007/s12035-024-04216-2] [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: 01/28/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024]
Abstract
Traumatic brain injury (TBI) is a highly severe form of trauma with complex series of reactions in brain tissue which ultimately results in neuronal damage. Previous studies proved that neuronal ferroptosis, which was induced by intracranial haemorrhage and other reasons, was one of the most primary causes of neuronal damage following TBI. However, the association between neuronal mechanical injury and ferroptosis in TBI and relevant treatments remain unclear. In the present study, we first demonstrated the occurrence of neuronal ferroptosis in the early stage of TBI and preliminarily elucidated that edaravone (EDA), a cerebroprotective agent that eliminates oxygen radicals, was able to inhibit ferroptosis induced by TBI. A cell scratching model was established in PC12 cells, and it was confirmed that mechanical injury induced ferroptosis in neurons at the early stage of TBI. Ferroptosis suppressor protein 1 (FSP1) plays a significant role in inhibiting ferroptosis, and we found that iFSP, a ferroptosis agonist which is capable to inhibit FSP1 pathway, attenuated the anti-ferroptosis effect of EDA. In conclusion, our results suggested that EDA inhibited neuronal ferroptosis induced by mechanical injury in the early phase of TBI by activating FSP1 pathway, which could provide evidence for future research on prevention and treatment of TBI.
Collapse
Affiliation(s)
- Haoyu Shi
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904th Hospital of PLA)/Fifth Clinical Medical College of Anhui Medical University, Wuxi, 214044, Jiangsu Province, China
| | - Libiao Song
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904th Hospital of PLA)/Fifth Clinical Medical College of Anhui Medical University, Wuxi, 214044, Jiangsu Province, China
| | - Yonghui Wu
- Department of Neurosurgery, The Second People's Hospital of Lu'an, Lu'an, 237000, Anhui Province, China
| | - Ruonan Shen
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904th Hospital of PLA)/Fifth Clinical Medical College of Anhui Medical University, Wuxi, 214044, Jiangsu Province, China
| | - Chenxu Zhang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904th Hospital of PLA)/Fifth Clinical Medical College of Anhui Medical University, Wuxi, 214044, Jiangsu Province, China
| | - Xingzhi Liao
- Department of Anaesthesiology, Wuxi Clinical College of Anhui Medical University (The 904th Hospital of PLA)/Fifth Clinical Medical College of Anhui Medical University, Wuxi, 214044, Jiangsu Province, China
| | - Qiuhong Wang
- Department of Ophthalmology, Wuxi Second Hospital Affiliated to Jiangnan University, Wuxi, 214002, Jiangsu Province, China
| | - Jie Zhu
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904th Hospital of PLA)/Fifth Clinical Medical College of Anhui Medical University, Wuxi, 214044, Jiangsu Province, China.
| |
Collapse
|
9
|
Wang YY, Li K, Wang JJ, Hua W, Liu Q, Sun YL, Qi JP, Song YJ. Bone marrow-derived mesenchymal stem cell-derived exosome-loaded miR-129-5p targets high-mobility group box 1 attenuates neurological-impairment after diabetic cerebral hemorrhage. World J Diabetes 2024; 15:1979-2001. [PMID: 39280179 PMCID: PMC11372641 DOI: 10.4239/wjd.v15.i9.1979] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Diabetic intracerebral hemorrhage (ICH) is a serious complication of diabetes. The role and mechanism of bone marrow mesenchymal stem cell (BMSC)-derived exosomes (BMSC-exo) in neuroinflammation post-ICH in patients with diabetes are unknown. In this study, we investigated the regulation of BMSC-exo on hyperglycemia-induced neuroinflammation. AIM To study the mechanism of BMSC-exo on nerve function damage after diabetes complicated with cerebral hemorrhage. METHODS BMSC-exo were isolated from mouse BMSC media. This was followed by transfection with microRNA-129-5p (miR-129-5p). BMSC-exo or miR-129-5p-overexpressing BMSC-exo were intravitreally injected into a diabetes mouse model with ICH for in vivo analyses and were cocultured with high glucose-affected BV2 cells for in vitro analyses. The dual luciferase test and RNA immunoprecipitation test verified the targeted binding relationship between miR-129-5p and high-mobility group box 1 (HMGB1). Quantitative polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay were conducted to assess the levels of some inflammation factors, such as HMGB1, interleukin 6, interleukin 1β, toll-like receptor 4, and tumor necrosis factor α. Brain water content, neural function deficit score, and Evans blue were used to measure the neural function of mice. RESULTS Our findings indicated that BMSC-exo can promote neuroinflammation and functional recovery. MicroRNA chip analysis of BMSC-exo identified miR-129-5p as the specific microRNA with a protective role in neuroinflammation. Overexpression of miR-129-5p in BMSC-exo reduced the inflammatory response and neurological impairment in comorbid diabetes and ICH cases. Furthermore, we found that miR-129-5p had a targeted binding relationship with HMGB1 mRNA. CONCLUSION We demonstrated that BMSC-exo can reduce the inflammatory response after ICH with diabetes, thereby improving the neurological function of the brain.
Collapse
Affiliation(s)
- Yue-Ying Wang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Ke Li
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Jia-Jun Wang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Wei Hua
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Qi Liu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yu-Lan Sun
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Ji-Ping Qi
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yue-Jia Song
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
10
|
Wang YY, Li K, Wang JJ, Hua W, Liu Q, Sun YL, Qi JP, Song YJ. Bone marrow-derived mesenchymal stem cell-derived exosome-loaded miR-129-5p targets high-mobility group box 1 attenuates neurological-impairment after diabetic cerebral hemorrhage. World J Diabetes 2024; 15:1978-2000. [DOI: 10.4239/wjd.v15.i9.1978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Diabetic intracerebral hemorrhage (ICH) is a serious complication of diabetes. The role and mechanism of bone marrow mesenchymal stem cell (BMSC)-derived exosomes (BMSC-exo) in neuroinflammation post-ICH in patients with diabetes are unknown. In this study, we investigated the regulation of BMSC-exo on hyperglycemia-induced neuroinflammation.
AIM To study the mechanism of BMSC-exo on nerve function damage after diabetes complicated with cerebral hemorrhage.
METHODS BMSC-exo were isolated from mouse BMSC media. This was followed by transfection with microRNA-129-5p (miR-129-5p). BMSC-exo or miR-129-5p-overexpressing BMSC-exo were intravitreally injected into a diabetes mouse model with ICH for in vivo analyses and were cocultured with high glucose-affected BV2 cells for in vitro analyses. The dual luciferase test and RNA immunoprecipitation test verified the targeted binding relationship between miR-129-5p and high-mobility group box 1 (HMGB1). Quantitative polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay were conducted to assess the levels of some inflammation factors, such as HMGB1, interleukin 6, interleukin 1β, toll-like receptor 4, and tumor necrosis factor α. Brain water content, neural function deficit score, and Evans blue were used to measure the neural function of mice.
RESULTS Our findings indicated that BMSC-exo can promote neuroinflammation and functional recovery. MicroRNA chip analysis of BMSC-exo identified miR-129-5p as the specific microRNA with a protective role in neuroinflammation. Overexpression of miR-129-5p in BMSC-exo reduced the inflammatory response and neurological impairment in comorbid diabetes and ICH cases. Furthermore, we found that miR-129-5p had a targeted binding relationship with HMGB1 mRNA.
CONCLUSION We demonstrated that BMSC-exo can reduce the inflammatory response after ICH with diabetes, thereby improving the neurological function of the brain.
Collapse
Affiliation(s)
- Yue-Ying Wang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Ke Li
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Jia-Jun Wang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Wei Hua
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Qi Liu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yu-Lan Sun
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Ji-Ping Qi
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yue-Jia Song
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
11
|
Li L, Liu X, Han C, Tian L, Wang Y, Han B. Ferroptosis in radiation-induced brain injury: roles and clinical implications. Biomed Eng Online 2024; 23:93. [PMID: 39261942 PMCID: PMC11389269 DOI: 10.1186/s12938-024-01288-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: 12/22/2023] [Accepted: 08/31/2024] [Indexed: 09/13/2024] Open
Abstract
Radiation-induced brain injury (RBI) presents a significant challenge for patients undergoing radiation therapy for head, neck, and intracranial tumors. This review aims to elucidate the role of ferroptosis in RBI and its therapeutic implications. Specifically, we explore how ferroptosis can enhance the sensitivity of tumor cells to radiation while also examining strategies to mitigate radiation-induced damage to normal brain tissues. By investigating the mechanisms through which radiation increases cellular reactive oxygen species (ROS) and initiates ferroptosis, we aim to develop targeted therapeutic strategies that maximize treatment efficacy and minimize neurotoxicity. The review highlights key regulatory factors in the ferroptosis pathway, including glutathione peroxidase 4 (GPX4), cystine/glutamate antiporter system Xc- (System Xc-), nuclear factor erythroid 2-related factor 2 (NRF2), Acyl-CoA synthetase long-chain family member 4 (ACSL4), and others, and their interactions in the context of RBI. Furthermore, we discuss the clinical implications of modulating ferroptosis in radiation therapy, emphasizing the potential for selective induction of ferroptosis in tumor cells and inhibition in healthy cells. The development of advanced diagnostic tools and therapeutic strategies targeting ferroptosis offers a promising avenue for enhancing the safety and efficacy of radiation therapy, underscoring the need for further research in this burgeoning field.
Collapse
Affiliation(s)
- Lifang Li
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Xia Liu
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Chunfeng Han
- Department of Pharmacy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Licheng Tian
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Yongzhi Wang
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Baolin Han
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China.
| |
Collapse
|
12
|
Xiao H, Bao X, Bai N, Zhu W, Saqirila S, Hu X, Bao Q, Baigude H. Synthesis of Lipidated Ligands and Formulation of Glia-Specific LNPs for RNAi-Mediated BBB Protection. J Med Chem 2024. [PMID: 39031092 DOI: 10.1021/acs.jmedchem.4c01176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2024]
Abstract
Pro-inflammatory polarization of microglia and astrocytes results in neuroinflammation and blood-brain barrier (BBB) disruption after a primary traumatic brain injury (TBI). Herein, we demonstrate that the dual-ligand functionalized lipid nanoparticles (AM31 LNPs) were actively and specifically internalized by microglia and astrocytes via mannose receptor (MR)- and adenosine receptor (AR)-mediated endocytosis, respectively, in a mouse model of TBI. Systemic administration of AM31 LNPs carrying siRNA against p65 resulted in internalization by the glial cells in the peri-infarct region and a robust knockdown of p65 at both mRNA and protein levels in these cells, leading to significant down-regulation of key pro-inflammatory cytokines and up-regulation of key anti-inflammatory cytokines. AM31 LNP-mediated silencing of p65 ameliorated TBI-induced BBB disruption. Our data proved that AM 31 LNP is a promising vehicle for RNA therapeutics for targeting microglia and astrocytes in neural disorder.
Collapse
Affiliation(s)
- Hai Xiao
- School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Xuemei Bao
- School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Nuomin Bai
- School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Wunile Zhu
- School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Saqirila Saqirila
- School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Xin Hu
- School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Qingming Bao
- School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Huricha Baigude
- School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| |
Collapse
|
13
|
Yang Y, Li Z, Fan X, Jiang C, Wang J, Rastegar-Kashkooli Y, Wang TJ, Wang J, Wang M, Cheng N, Yuan X, Chen X, Jiang B, Wang J. Nanozymes: Potential Therapies for Reactive Oxygen Species Overproduction and Inflammation in Ischemic Stroke and Traumatic Brain Injury. ACS NANO 2024; 18:16450-16467. [PMID: 38897929 DOI: 10.1021/acsnano.4c03425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Nanozymes, which can selectively scavenge reactive oxygen species (ROS), have recently emerged as promising candidates for treating ischemic stroke and traumatic brain injury (TBI) in preclinical models. ROS overproduction during the early phase of these diseases leads to oxidative brain damage, which has been a major cause of mortality worldwide. However, the clinical application of ROS-scavenging enzymes is limited by their short in vivo half-life and inability to cross the blood-brain barrier. Nanozymes, which mimic the catalytic function of natural enzymes, have several advantages, including cost-effectiveness, high stability, and easy storage. These advantages render them superior to natural enzymes for disease diagnosis and therapeutic interventions. This review highlights recent advancements in nanozyme applications for ischemic stroke and TBI, emphasizing their potential to mitigate the detrimental effect of ROS overproduction, oxidative brain damage, inflammation, and blood-brain barrier compromise. Therefore, nanozymes represent a promising treatment modality for ROS overproduction conditions in future medical practices.
Collapse
Affiliation(s)
- Yunfan Yang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Zixiang Li
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
- School of International Education, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Tom J Wang
- Program in Behavioral Biology, The Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Nannan Cheng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Xiqian Yuan
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| |
Collapse
|
14
|
Zhang M, Han X, Yan L, Fu Y, Kou H, Shang C, Wang J, Liu H, Jiang C, Wang J, Cheng T. Inflammatory response in traumatic brain and spinal cord injury: The role of XCL1-XCR1 axis and T cells. CNS Neurosci Ther 2024; 30:e14781. [PMID: 38887195 PMCID: PMC11183917 DOI: 10.1111/cns.14781] [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: 01/12/2024] [Revised: 04/29/2024] [Accepted: 05/11/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) and spinal cord injury (SCI) are acquired injuries to the central nervous system (CNS) caused by external forces that cause temporary or permanent sensory and motor impairments and the potential for long-term disability or even death. These conditions currently lack effective treatments and impose substantial physical, social, and economic burdens on millions of people and families worldwide. TBI and SCI involve intricate pathological mechanisms, and the inflammatory response contributes significantly to secondary injury in TBI and SCI. It plays a crucial role in prolonging the post-CNS trauma period and becomes a focal point for a potential therapeutic intervention. Previous research on the inflammatory response has traditionally concentrated on glial cells, such as astrocytes and microglia. However, increasing evidence highlights the crucial involvement of lymphocytes in the inflammatory response to CNS injury, particularly CD8+ T cells and NK cells, along with their downstream XCL1-XCR1 axis. OBJECTIVE This review aims to provide an overview of the role of the XCL1-XCR1 axis and the T-cell response in inflammation caused by TBI and SCI and identify potential targets for therapy. METHODS We conducted a comprehensive search of PubMed and Web of Science using relevant keywords related to the XCL1-XCR1 axis, T-cell response, TBI, and SCI. RESULTS This study examines the upstream and downstream pathways involved in inflammation caused by TBI and SCI, including interleukin-15 (IL-15), interleukin-12 (IL-12), CD8+ T cells, CD4+ T cells, NK cells, XCL1, XCR1+ dendritic cells, interferon-gamma (IFN-γ), helper T0 cells (Th0 cells), helper T1 cells (Th1 cells), and helper T17 cells (Th17 cells). We describe their proinflammatory effect in TBI and SCI. CONCLUSIONS The findings suggest that the XCL1-XCR1 axis and the T-cell response have great potential for preclinical investigations and treatments for TBI and SCI.
Collapse
Affiliation(s)
- Mingkang Zhang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiaonan Han
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Liyan Yan
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yikun Fu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongwei Kou
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chunfeng Shang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Hongjian Liu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chao Jiang
- Department of NeurologyPeople's Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Tian Cheng
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
15
|
Zhang L, Bai W, Peng Y, Lin Y, Tian M. Human umbilical cord mesenchymal stem cell-derived exosomes provide neuroprotection in traumatic brain injury through the lncRNA TUBB6/Nrf2 pathway. Brain Res 2024; 1824:148689. [PMID: 38030103 DOI: 10.1016/j.brainres.2023.148689] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/16/2023] [Accepted: 11/25/2023] [Indexed: 12/01/2023]
Abstract
Recently, human umbilical cord mesenchymal stem cell (HucMSC) is a new focus of research in neurological diseases, and the beneficial effect of HucMSC is mediated by paracrine factors which are transported by exosome. Our previous study has shown that HucMSC-derived exosome could provide neuroprotection after traumatic brain injury (TBI). However, the underlying mechanisms were not fully understood. In the present study, we found that administration of exosome suppressed TBI-induced inflammation and ferroptosis. In addition, exosome activated the long non-coding ribonucleic acid (lncRNA) TUBB6/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway after TBI. However, exosome partly failed to provide neuroprotection following TBI when TUBB6 was knockdown. Importantly, exosome treatment also decreased neuron cell death, suppressed inflammation, inhibited ferroptosis and activated the lncRNA TUBB6/Nrf2 pathway after TBI in vitro. Taken together, our results provided the first evidence that HucMSC-derived exosome played a key role in neuroprotection after TBI through the lncRNA TUBB6/Nrf2 pathway.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Wanshan Bai
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Yaonan Peng
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Yixing Lin
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Mi Tian
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, PR China.
| |
Collapse
|
16
|
Li N, Wang R, Ai X, Guo J, Bai Y, Guo X, Zhang R, Du X, Chen J, Li H. Electroacupuncture Inhibits Neural Ferroptosis in Rat Model of Traumatic Brain Injury via Activating System Xc -/GSH/GPX4 Axis. Curr Neurovasc Res 2024; 21:86-100. [PMID: 38629369 DOI: 10.2174/0115672026297775240405073502] [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: 12/07/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Ferroptosis is an iron-dependent regulating programmed cell death discovered recently that has been receiving much attention in traumatic brain injury (TBI). xCT, a major functional subunit of Cystine/glutamic acid reverse transporter (System Xc-), promotes cystine intake and glutathione biosynthesis, thereby protecting against oxidative stress and ferroptosis. OBJECTIVE The intention of this research was to verify the hypothesis that electroacupuncture (EA) exerted an anti-ferroptosis effect via an increase in the expression of xCT and activation of the System Xc-/GSH/GPX4 axis in cortical neurons of TBI rats. METHODS After the TBI rat model was prepared, animals received EA treatment at GV20, GV26, ST36 and PC6, for 15 min. The xCT inhibitor Sulfasalazine (SSZ) was administered 2h prior to model being prepared. The degree of neurological impairment was evaluated by means of TUNEL staining and the modified neurological severity score (mNSS). Specific indicators of ferroptosis (Ultrastructure of mitochondria, Iron and ROS) were detected by transmission electron microscopy (TEM), Prussian blue staining (Perls stain) and flow cytometry (FCM), respectively. GSH synthesis and metabolism-related factors in the content of the cerebral cortex were detected by an assay kit. Real-time quantitative PCR (RT-QPCR), Western blot (WB), and immunofluorescence (IF) were used for detecting the expression of System Xc-/GSH/GPX4 axisrelated proteins in injured cerebral cortex tissues. RESULTS EA successfully relieved nerve damage within 7 days after TBI, significantly inhibited neuronal ferroptosis, upregulated the expression of xCT and System Xc-/GSH/GPX4 axis forward protein and promoted glutathione (GSH) synthesis and metabolism in the injured area of the cerebral cortex. However, aggravation of nerve damage and increased ferroptosis effect were found in TBI rats injected with xCT inhibitors. CONCLUSIONS EA inhibits neuronal ferroptosis by up-regulated xCT expression and by activating System Xc-/GSH/GPX4 axis after TBI, confirming the relevant theories regarding the EA effect in treating TBI and providing theoretical support for clinical practice.
Collapse
Affiliation(s)
- Na Li
- School of Acupuncture-Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi'an, Shaanxi, 712046, China
| | - Ruihui Wang
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi'an, Shaanxi, 712046, China
| | - Xia Ai
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi'an, Shaanxi, 712046, China
| | - Jie Guo
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi'an, Shaanxi, 712046, China
| | - Yuwang Bai
- Department of Pneumology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, 710001, China
| | - Xinrong Guo
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi'an, Shaanxi, 712046, China
| | - Rongchao Zhang
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi'an, Shaanxi, 712046, China
| | - Xu Du
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi'an, Shaanxi, 712046, China
| | - Jingxuan Chen
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi'an, Shaanxi, 712046, China
| | - Hua Li
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi'an, Shaanxi, 712046, China
| |
Collapse
|
17
|
Alqahtani F, Mohamed Ali YS, Almutairi MM, Alotaibi AF, Imran I, Alshammari MA, Alshememry AK, AlSharari SD, Albekairi TH. Therapeutic benefits of quercetin in traumatic brain injury model exposed to cigarette smoke. Saudi Pharm J 2024; 32:101895. [PMID: 38226352 PMCID: PMC10788629 DOI: 10.1016/j.jsps.2023.101895] [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: 10/22/2023] [Accepted: 12/03/2023] [Indexed: 01/17/2024] Open
Abstract
Scientific evidences reported the deleterious effect of cigarette smoking or passive smoking on brain health particularly cognitive functions, blood-brain barrier (BBB) permeability, up-regulation of inflammatory cascades, and depletion of the antioxidant system. These combined effects become more progressive in the events of stroke, traumatic brain injury (TBI), and many other neurodegenerative diseases. In the current study, we investigated the long-term administered therapeutic potential of quercetin in ameliorating the deleterious neurobiological consequences of chronic tobacco smoke exposure in TBI mice. After exposure to 21 days of cigarette smoke and treatment with 50 mg/kg of quercetin, C57BL/6 mice were challenged for the induction of TBI by the weight drop method. Subsequently, a battery of behavioral tests and immunohistochemical analyses revealed the beneficial effect of quercetin on the locomotive and cognitive function of TBI + smoked group mice (p < 0.05 vs control sham). Immunohistochemistry analysis (Nrf2, HO-1, NFkB, caspase 3) demonstrated a marked protection after 21 days of quercetin treatment in the chronic tobacco smoking group possibly by up-regulation of antioxidant pathways, and decreased apoptosis. In conclusion, our findings support the therapeutic effectiveness of quercetin in partly protecting the central neurological functions that become aberrantly impaired in combined habitual cigarette-smoking individuals impacted with TBI.
Collapse
Affiliation(s)
- Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yousif S. Mohamed Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah F. Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah K. Alshememry
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shakir D. AlSharari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Thamer H. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
18
|
Shang J, Li W, Zhang H, Wang W, Liu N, Gao D, Wang F, Yan X, Gao C, Sun R, Zhang H, Ma K, Shao F, Zhang J. C-kit controls blood-brain barrier permeability by regulating caveolae-mediated transcytosis after chronic cerebral hypoperfusion. Biomed Pharmacother 2024; 170:115778. [PMID: 38141279 DOI: 10.1016/j.biopha.2023.115778] [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: 07/12/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 12/25/2023] Open
Abstract
Blood-brain barrier (BBB) dysfunction plays a pivotal role in the pathology of chronic cerebral hypoperfusion (CCH)-related neurodegenerative diseases. Continuous endothelial cells (EC) that line the blood vessels of the brain are important components of the BBB to strictly control the flow of substances and maintain the homeostatic environment of the brain. However, the molecular mechanisms from the perspective of EC-induced BBB dysfunction after CCH are largely unknown. In this study, the BBB function was assessed using immunostaining and transmission electron microscopy. The EC dysfunction profile was screened by using EC enrichment followed by RNA sequencing. After identified the key EC dysfunction factor, C-kit, we used the C-kit inhibition drug (imatinib) and C-kit down-regulation method (AAV-BR1-C-kit shRNA) to verify the role of C-kit on BBB integrity and EC transcytosis after CCH. Furthermore, we also activated C-kit with stem cell factor (SCF) to observe the effects of C-kit on BBB following CCH. We explored that macromolecular proteins entered the brain mainly through EC transcytosis after CCH and caused neuronal loss. Additionally, we identified receptor tyrosine kinase C-kit as a key EC dysfunction molecule. Furthermore, the pharmacological inhibition of C-kit with imatinib counteracted BBB leakage by reducing caveolae-mediated transcytosis. Moreover, treatment with AAV-BR1-C-kit shRNA, which targets brain EC to inhibit C-kit expression, also ameliorated BBB leakage by reducing caveolae-mediated transcytosis. Furthermore, the SCF increased the permeability of the BBB by actively increasing caveolae-mediated transcytosis. This study provides evidence that C-kit is a key BBB permeability regulator through caveolae-mediated transcytosis in EC after CCH.
Collapse
Affiliation(s)
- Junkui Shang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Wei Li
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Huiwen Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Wan Wang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Ning Liu
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Dandan Gao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430072, China
| | - Fengyu Wang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Xi Yan
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Chenhao Gao
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Ruihua Sun
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Haohan Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Kai Ma
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Fengmin Shao
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China; Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, China.
| | - Jiewen Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| |
Collapse
|
19
|
Nie L, He J, Wang J, Wang R, Huang L, Jia L, Kim YT, Bhawal UK, Fan X, Zille M, Jiang C, Chen X, Wang J. Environmental Enrichment for Stroke and Traumatic Brain Injury: Mechanisms and Translational Implications. Compr Physiol 2023; 14:5291-5323. [PMID: 38158368 DOI: 10.1002/cphy.c230007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Acquired brain injuries, such as ischemic stroke, intracerebral hemorrhage (ICH), and traumatic brain injury (TBI), can cause severe neurologic damage and even death. Unfortunately, currently, there are no effective and safe treatments to reduce the high disability and mortality rates associated with these brain injuries. However, environmental enrichment (EE) is an emerging approach to treating and rehabilitating acquired brain injuries by promoting motor, sensory, and social stimulation. Multiple preclinical studies have shown that EE benefits functional recovery, including improved motor and cognitive function and psychological benefits mediated by complex protective signaling pathways. This article provides an overview of the enriched environment protocols used in animal models of ischemic stroke, ICH, and TBI, as well as relevant clinical studies, with a particular focus on ischemic stroke. Additionally, we explored studies of animals with stroke and TBI exposed to EE alone or in combination with multiple drugs and other rehabilitation modalities. Finally, we discuss the potential clinical applications of EE in future brain rehabilitation therapy and the molecular and cellular changes caused by EE in rodents with stroke or TBI. This article aims to advance preclinical and clinical research on EE rehabilitation therapy for acquired brain injury. © 2024 American Physiological Society. Compr Physiol 14:5291-5323, 2024.
Collapse
Affiliation(s)
- Luwei Nie
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxin He
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory for Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ruike Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Lin Jia
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
- Department of Food Biotechnology, Korea University of Science & Technology, Daejeon, Republic of Korea
| | - Ujjal K Bhawal
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, Japan
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
20
|
Wang Y, Hu J, Wu S, Fleishman JS, Li Y, Xu Y, Zou W, Wang J, Feng Y, Chen J, Wang H. Targeting epigenetic and posttranslational modifications regulating ferroptosis for the treatment of diseases. Signal Transduct Target Ther 2023; 8:449. [PMID: 38072908 PMCID: PMC10711040 DOI: 10.1038/s41392-023-01720-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/16/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Ferroptosis, a unique modality of cell death with mechanistic and morphological differences from other cell death modes, plays a pivotal role in regulating tumorigenesis and offers a new opportunity for modulating anticancer drug resistance. Aberrant epigenetic modifications and posttranslational modifications (PTMs) promote anticancer drug resistance, cancer progression, and metastasis. Accumulating studies indicate that epigenetic modifications can transcriptionally and translationally determine cancer cell vulnerability to ferroptosis and that ferroptosis functions as a driver in nervous system diseases (NSDs), cardiovascular diseases (CVDs), liver diseases, lung diseases, and kidney diseases. In this review, we first summarize the core molecular mechanisms of ferroptosis. Then, the roles of epigenetic processes, including histone PTMs, DNA methylation, and noncoding RNA regulation and PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, methylation, and ADP-ribosylation, are concisely discussed. The roles of epigenetic modifications and PTMs in ferroptosis regulation in the genesis of diseases, including cancers, NSD, CVDs, liver diseases, lung diseases, and kidney diseases, as well as the application of epigenetic and PTM modulators in the therapy of these diseases, are then discussed in detail. Elucidating the mechanisms of ferroptosis regulation mediated by epigenetic modifications and PTMs in cancer and other diseases will facilitate the development of promising combination therapeutic regimens containing epigenetic or PTM-targeting agents and ferroptosis inducers that can be used to overcome chemotherapeutic resistance in cancer and could be used to prevent other diseases. In addition, these mechanisms highlight potential therapeutic approaches to overcome chemoresistance in cancer or halt the genesis of other diseases.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jing Hu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300060, PR China
| | - Shuang Wu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, PR China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yulin Li
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Yinshi Xu
- Department of Outpatient, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Wailong Zou
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Yukuan Feng
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China.
| | - Hongquan Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| |
Collapse
|
21
|
Yan L, Han X, Zhang M, Fu Y, Yang F, Li Q, Cheng T. Integrative analysis of TBI data reveals Lgmn as a key player in immune cell-mediated ferroptosis. BMC Genomics 2023; 24:747. [PMID: 38057699 DOI: 10.1186/s12864-023-09842-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/24/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a central nervous system disease caused by external trauma, which has complex pathological and physiological mechanisms. The aim of this study was to explore the correlation between immune cell infiltration and ferroptosis post-TBI. METHODS This study utilized the GEO database to download TBI data and performed differentially expressed genes (DEGs) and ferroptosis-related differentially expressed genes (FRDEGs) analysis. DEGs were further analyzed for enrichment using the DAVID 6.8. Immunoinfiltration cell analysis was performed using the ssGSEA package and the Timer2.0 tool. The WGCNA analysis was then used to explore the gene modules in the data set associated with differential expression of immune cell infiltration and to identify the hub genes. The tidyverse package and corrplot package were used to calculate the correlations between hub genes and immune cell infiltration and ferroptosis-marker genes. The miRDB and TargetScan databases were used to predict complementary miRNAs for the Hub genes selected from the WGCNA analysis, and the DIANA-LncBasev3 tool was used to identify target lncRNAs for the miRNAs, constructing an mRNA-miRNA-lncRNA regulatory network. RESULTS A total of 320 DEGs and 21 FRDEGs were identified in GSE128543. GO and KEGG analyses showed that the DEGs after TBI were primarily associated with inflammation and immune response. Xcell and ssGSEA immune infiltration cell analysis showed significant infiltration of T cell CD4+ central memory, T cell CD4+ Th2, B cell memory, B cell naive, monocyte, macrophage, and myeloid dendritic cell activated. The WGCNA analysis identified two modules associated with differentially expressed immune cells and identified Lgmn as a hub gene associated with immune infiltrating cells. Lgmn showed significant correlation with immune cells and ferroptosis-marker genes, including Gpx4, Hspb1, Nfe2l2, Ptgs2, Fth1, and Tfrc. Finally, an mRNA-miRNA-lncRNA regulatory network was constructed using Lgmn. CONCLUSION Our results indicate that there is a certain correlation between ferroptosis and immune infiltrating cells in brain tissue after TBI, and that Lgmn plays an important role in this process.
Collapse
Affiliation(s)
- Liyan Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiaonan Han
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Mingkang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yikun Fu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Tian Cheng
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
22
|
Liu Y, Zhao Z, Guo J, Ma Y, Li J, Ji H, Chen Z, Zheng J. Anacardic acid improves neurological deficits in traumatic brain injury by anti-ferroptosis and anti-inflammation. Exp Neurol 2023; 370:114568. [PMID: 37820939 DOI: 10.1016/j.expneurol.2023.114568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/23/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is an important cause of disability and death. TBI leads to multiple forms of nerve cell death including ferroptosis due to iron-dependent lipid peroxidation. Anacardic acid (AA) is a natural component extracted from cashew nut shells, which has been reported to have neuroprotective effects in traumatic brain injury. We investigated whether AA has an anti-ferroptosis effect in TBI. METHODS We used the Feeney free-fall impact method to construct a TBI model to investigate the effect of AA on ferroptosis caused by TBI, in which Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, served as a positive control group. We first identified the therapeutic effect of AA on TBI through modified neurological severity score (mNSS) and determined the appropriate concentration. Secondly, we investigated the effect of AA on the expression level of the key protein of ferroptosis by Western blotting and immunohistochemistry. Then the effect of AA on nerve tissue injury and nerve function improvement was verified. Finally, enzym-linked immunosorbent assay (ELISA) was used to verify that AA could reduce inflammation after TBI. RESULTS We found the intensely inhibitory effect of AA on ferroptosis, which is in parallel with the results obtained after Fer-1 treatment. In addition, AA and Fer-1 mitigated TBI-mediated tissue defects, destruction of the blood-brain barrier, and neurodegeneration. Novel object recognition (NOR), mNSS and water maze test showed that AA could significantly reduce the impairment of neural function and behavioral cognitive ability caused by TBI. Finally, we also demonstrated that AA has not only an anti-ferroptosis effect, but also an anti-inflammation effect. CONCLUSIONS AA can reduce the neurological impairment and behavioral cognitive impairment caused by TBI through the dual effect of anti-ferroptosis and anti-inflammation.
Collapse
Affiliation(s)
- Yu Liu
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China; Xuzhou Medical University, Xuzhou 221000, China
| | - Zongren Zhao
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China
| | - Jianqiang Guo
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China; Xuzhou Medical University, Xuzhou 221000, China
| | - Yuanhao Ma
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China; Xuzhou Medical University, Xuzhou 221000, China
| | - Jing Li
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China
| | - Huanhuan Ji
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China
| | - Zhongjun Chen
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China
| | - Jinyu Zheng
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China.
| |
Collapse
|
23
|
Balena T, Lillis K, Rahmati N, Bahari F, Dzhala V, Berdichevsky E, Staley K. A Dynamic Balance between Neuronal Death and Clearance in an in Vitro Model of Acute Brain Injury. J Neurosci 2023; 43:6084-6107. [PMID: 37527922 PMCID: PMC10451151 DOI: 10.1523/jneurosci.0436-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/15/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
In in vitro models of acute brain injury, neuronal death may overwhelm the capacity for microglial phagocytosis, creating a queue of dying neurons awaiting clearance. Neurons undergoing programmed cell death are in this queue, and are the most visible and frequently quantified measure of neuronal death after injury. However, the size of this queue should be equally sensitive to changes in neuronal death and the rate of phagocytosis. Using rodent organotypic hippocampal slice cultures as a model of acute perinatal brain injury, serial imaging demonstrated that the capacity for microglial phagocytosis of dying neurons was overwhelmed for 2 weeks. Altering phagocytosis rates (e.g., by changing the number of microglia) dramatically changed the number of visibly dying neurons. Similar effects were generated when the visibility of dying neurons was altered by changing the membrane permeability for stains that label dying neurons. Canonically neuroprotective interventions, such as seizure blockade, and neurotoxic maneuvers, such as perinatal ethanol exposure, were mediated by effects on microglial activity and the membrane permeability of neurons undergoing programmed cell death. These canonically neuroprotective and neurotoxic interventions had either no or opposing effects on healthy surviving neurons identified by the ongoing expression of transgenic fluorescent proteins.SIGNIFICANCE STATEMENT In in vitro models of acute brain injury, microglial phagocytosis is overwhelmed by the number of dying cells. Under these conditions, the assumptions on which assays for neuroprotective and neurotoxic effects are based are no longer valid. Thus, longitudinal assays of healthy cells, such as serial assessment of the fluorescence emission of transgenically expressed proteins, provide more accurate estimates of cell death than do single-time point anatomic or biochemical assays of the number of dying neurons. More accurate estimates of death rates in vitro will increase the translatability of preclinical studies of neuroprotection and neurotoxicity.
Collapse
Affiliation(s)
- Trevor Balena
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kyle Lillis
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Negah Rahmati
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Fatemeh Bahari
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Volodymyr Dzhala
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Eugene Berdichevsky
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania 18015
| | - Kevin Staley
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| |
Collapse
|
24
|
Viktorinova A. Future Perspectives of Oxytosis/Ferroptosis Research in Neurodegeneration Diseases. Cell Mol Neurobiol 2023; 43:2761-2768. [PMID: 37093436 PMCID: PMC11410136 DOI: 10.1007/s10571-023-01353-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/14/2023] [Indexed: 04/25/2023]
Abstract
The current report briefly summarizes the existing hypotheses and relevant evidence of oxytosis/ferroptosis-mediated cell death and outlines future perspectives of neurodegeneration research. Furthermore, it highlights the potential application of specific markers (e.g., activators, inhibitors, redox modulators, antioxidants, iron chelators) in the study of regulatory mechanisms of oxytosis/ferroptosis. It appears that these markers may be a suitable option for experimental investigations targeting key pathways of oxytosis/ferroptosis, such as the inhibition of the cystine/glutamate antiporter/glutathione/glutathione peroxidase 4 axis, glutamate oxidative toxicity, glutathione depletion, iron dyshomeostasis, iron-mediated lipid peroxidation, and others. From a clinical perspective, an innovative research approach to investigate the oxytosis/ferroptosis pathways in cells of the central nervous system and their relationship to neurodegenerative diseases is desirable. It is necessary to expand the existing knowledge about the molecular mechanisms of neurodegenerative diseases and to provide innovative diagnostic procedures to prevent their progression, as well as to develop effective neuroprotective treatment. The importance of preclinical studies focused predominantly on oxytosis/ferroptosis inhibitors (iron chelators or lipoxygenase inhibitors and lipophilic antioxidants) that could chelate iron or inhibit lipid peroxidation is also discussed. Specifically, this targeted inhibition of neuronal death could represent a potential therapeutic strategy for some neurodegenerative diseases.
Collapse
Affiliation(s)
- Alena Viktorinova
- Faculty of Medicine, Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Comenius University in Bratislava, Sasinkova 2, 811 08, Bratislava, Slovakia.
| |
Collapse
|
25
|
Zhang Q, Fan X, Zhang X, Ju S. Ferroptosis in tumors and its relationship to other programmed cell death: role of non-coding RNAs. J Transl Med 2023; 21:514. [PMID: 37516888 PMCID: PMC10387214 DOI: 10.1186/s12967-023-04370-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023] Open
Abstract
Programmed cell death (PCD) plays an important role in many aspects of individual development, maintenance of body homeostasis and pathological processes. Ferroptosis is a novel form of PCD characterized by the accumulation of iron-dependent lipid peroxides resulting in lethal cell damage. It contributes to tumor progression in an apoptosis-independent manner. In recent years, an increasing number of non-coding RNAs (ncRNAs) have been demonstrated to mediate the biological process of ferroptosis, hence impacting carcinogenesis, progression, drug resistance, and prognosis. However, the clear regulatory mechanism for this phenomenon remains poorly understood. Moreover, ferroptosis does not usually exist independently. Its interaction with PCD, like apoptosis, necroptosis, autophagy, pyroptosis, and cuproptosis, to destroy cells appears to exist. Furthermore, ncRNA seems to be involved. Here, we review the mechanisms by which ferroptosis occurs, dissect its relationship with other forms of death, summarize the key regulatory roles played by ncRNAs, raise relevant questions and predict possible barriers to its application in the clinic, offering new ideas for targeted tumour therapy.
Collapse
Affiliation(s)
- Qi Zhang
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xinfeng Fan
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xinyu Zhang
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- Department of Medical School of Nantong University, No.19, Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Shaoqing Ju
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, No.20, Xisi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
26
|
Penolazzi L, Straudi S, Lamberti N, Lambertini E, Bianchini C, Manfredini F, Piva R. Clinically-driven design of novel methods of investigation on skeletal health status in neurological disorders. The case of the traumatic brain injuries. Front Neurol 2023; 14:1176420. [PMID: 37265470 PMCID: PMC10230040 DOI: 10.3389/fneur.2023.1176420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/24/2023] [Indexed: 06/03/2023] Open
|
27
|
Sivandzade F, Alqahtani F, Dhaibar H, Cruz-Topete D, Cucullo L. Antidiabetic Drugs Can Reduce the Harmful Impact of Chronic Smoking on Post-Traumatic Brain Injuries. Int J Mol Sci 2023; 24:6219. [PMID: 37047198 PMCID: PMC10093862 DOI: 10.3390/ijms24076219] [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: 03/04/2023] [Revised: 03/18/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Traumatic Brain Injury (TBI) is a primary cause of cerebrovascular and neurological disorders worldwide. The current scientific researchers believe that premorbid conditions such as tobacco smoking (TS) can exacerbate post-TBI brain injury and negatively affect recovery. This is related to vascular endothelial dysfunction resulting from the exposure to TS-released reactive oxygen species (ROS), nicotine, and oxidative stress (OS) stimuli impacting the blood-brain barrier (BBB) endothelium. Interestingly, these pathogenic modulators of BBB impairment are similar to those associated with hyperglycemia. Antidiabetic drugs such as metformin (MF) and rosiglitazone (RSG) were shown to prevent/reduce BBB damage promoted by chronic TS exposure. Thus, using in vivo approaches, we evaluated the effectiveness of post-TBI treatment with MF or RSG to reduce the TS-enhancement of BBB damage and brain injury after TBI. For this purpose, we employed an in vivo weight-drop TBI model using male C57BL/6J mice chronically exposed to TS with and without post-traumatic treatment with MF or RSG. Our results revealed that these antidiabetic drugs counteracted TS-promoted downregulation of nuclear factor erythroid 2-related factor 2 (NRF2) expression and concomitantly dampened TS-enhanced OS, inflammation, and loss of BBB integrity following TBI. In conclusion, our findings suggest that MF and RSG could reduce the harmful impact of chronic smoking on post-traumatic brain injuries.
Collapse
Affiliation(s)
- Farzane Sivandzade
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11362, Saudi Arabia
| | - Hemangini Dhaibar
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Diana Cruz-Topete
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Luca Cucullo
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
28
|
Wang Y, Wu J, Wang J, He L, Lai H, Zhang T, Wang X, Li W. Mitochondrial oxidative stress in brain microvascular endothelial cells: Triggering blood-brain barrier disruption. Mitochondrion 2023; 69:71-82. [PMID: 36709855 DOI: 10.1016/j.mito.2023.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/02/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Blood-brain barrier disruption plays an important role in central nervous system diseases. This review provides information on the role of mitochondrial oxidative stress in brain microvascular endothelial cells in cellular dysfunction, the disruption of intercellular junctions, transporter dysfunction, abnormal angiogenesis, neurovascular decoupling, and the involvement and aggravation of vascular inflammation and illustrates related molecular mechanisms. In addition, recent drug and nondrug therapies targeting cerebral vascular endothelial cell mitochondria to repair the blood-brain barrier are discussed. This review shows that mitochondrial oxidative stress disorder in brain microvascular endothelial cells plays a key role in the occurrence and development of blood-brain barrier damage and may be critical in various pathological mechanisms of blood-brain barrier damage. These new findings suggest a potential new strategy for the treatment of central nervous system diseases through mitochondrial modulation of cerebral vascular endothelial cells.
Collapse
Affiliation(s)
- Yi Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jing Wu
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jiexin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Linxi He
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Han Lai
- School of Foreign Languages, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Tian Zhang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Xin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Weihong Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| |
Collapse
|
29
|
Balena T, Lillis K, Rahmati N, Bahari F, Dzhala V, Berdichevsky E, Staley K. A dynamic balance between neuronal death and clearance after acute brain injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528332. [PMID: 36824708 PMCID: PMC9948967 DOI: 10.1101/2023.02.14.528332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
After acute brain injury, neuronal apoptosis may overwhelm the capacity for microglial phagocytosis, creating a queue of dying neurons awaiting clearance. The size of this queue should be equally sensitive to changes in neuronal death and the rate of phagocytosis. Using rodent organotypic hippocampal slice cultures as a model of acute perinatal brain injury, serial imaging demonstrated that the capacity for microglial phagocytosis of dying neurons was overwhelmed for two weeks. Altering phagocytosis rates, e.g. by changing the number of microglia, dramatically changed the number of visibly dying neurons. Similar effects were generated when the visibility of dying neurons was altered by changing the membrane permeability for vital stains. Canonically neuroprotective interventions such as seizure blockade and neurotoxic maneuvers such as perinatal ethanol exposure were mediated by effects on microglial activity and the membrane permeability of apoptotic neurons, and had either no or opposing effects on healthy surviving neurons. Significance After acute brain injury, microglial phagocytosis is overwhelmed by the number of dying cells. Under these conditions, the assumptions on which assays for neuroprotective and neurotoxic effects are based are no longer valid. Thus longitudinal assays of healthy cells, such as assessment of the fluorescence emission of transgenically-expressed proteins, provide more accurate estimates of cell death than do single-time-point anatomical or biochemical assays. More accurate estimates of death rates will increase the translatability of preclinical studies of neuroprotection and neurotoxicity.
Collapse
|
30
|
Gareev I, Beylerli O, Liang Y, Lu E, Ilyasova T, Sufianov A, Sufianova G, Shi H, Ahmad A, Yang G. The Role of Mitochondria-Targeting miRNAs in Intracerebral Hemorrhage. Curr Neuropharmacol 2023; 21:1065-1080. [PMID: 35524670 PMCID: PMC10286585 DOI: 10.2174/1570159x20666220507021445] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 04/02/2022] [Accepted: 04/24/2022] [Indexed: 11/22/2022] Open
Abstract
Non-traumatic intracerebral hemorrhage (ICH) is the most common type of hemorrhagic stroke, most often occurring between the ages of 45 and 60. Arterial hypertension (AH) is most often the cause of ICH, followed by atherosclerosis, blood diseases, inflammatory changes in cerebral vessels, intoxication and vitamin deficiencies. Cerebral hemorrhage can occur by diapedesis or as a result of a ruptured vessel. AH is difficult to treat, requires surgery and can lead to disability or death. One of the important directions in the study of the pathogenesis of ICH is mitochondrial dysfunction and its regulation. The key role of mitochondrial dysfunction in AH and atherosclerosis, as well as in the development of brain damage after hemorrhage, has been acknowledged. MicroRNAs (miRNAs) are a class of non-coding RNAs (about 18-22 nucleotides) that regulate a variety of biological processes including cell differentiation, proliferation, apoptosis, etc., primarily through gene repression. There is growing evidence to support dysregulated miRNAs in various cardiovascular diseases, including ICH. Further, the realization of miRNAs within mitochondrial compartment has challenged the traditional knowledge of signaling pathways involved in the regulatory network of cardiovascular diseases. However, the role of miRNAs in mitochondrial dysfunction for ICH is still under-appreciated, with comparatively much lesser studies and investigations reported, than those in other cardiovascular diseases. In this review, we summarize the up-to-date findings on the published role miRNAs in mitochondrial function for ICH, and the potential use of miRNAs in clinical settings, such as potential therapeutic targets and non-invasive diagnostic/prognostic biomarker tools.
Collapse
Affiliation(s)
- Ilgiz Gareev
- Federal Centre of Neurosurgery, Tyumen, Russia
- Рeoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Ozal Beylerli
- Federal Centre of Neurosurgery, Tyumen, Russia
- Рeoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Yanchao Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Institute of Brain Science, Harbin Medical University, Harbin, 150001, China
| | - Enzhou Lu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Institute of Brain Science, Harbin Medical University, Harbin, 150001, China
| | - Tatiana Ilyasova
- Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Albert Sufianov
- Federal Centre of Neurosurgery, Tyumen, Russia
- Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Рeoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Galina Sufianova
- Department of Pharmacology, Tyumen State Medical University, Tyumen, Russia
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Institute of Brain Science, Harbin Medical University, Harbin, 150001, China
| | - Aamir Ahmad
- Interim Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Institute of Brain Science, Harbin Medical University, Harbin, 150001, China
| |
Collapse
|
31
|
Xu XJ, Liu BY, Dong JQ, Ge QQ, Lu SH, Yang MS, Zhuang Y, Zhang B, Niu F. Tandem Mass Tag-based proteomics analysis reveals the vital role of inflammation in traumatic brain injury in a mouse model. Neural Regen Res 2023. [PMID: 35799536 PMCID: PMC9241417 DOI: 10.4103/1673-5374.343886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
32
|
Song D, Yeh CT, Wang J, Guo F. Perspectives on the mechanism of pyroptosis after intracerebral hemorrhage. Front Immunol 2022; 13:989503. [PMID: 36131917 PMCID: PMC9484305 DOI: 10.3389/fimmu.2022.989503] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/17/2022] [Indexed: 12/18/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a highly harmful neurological disorder with high rates of mortality, disability, and recurrence. However, effective therapies are not currently available. Secondary immune injury and cell death are the leading causes of brain injury and a poor prognosis. Pyroptosis is a recently discovered form of programmed cell death that differs from apoptosis and necrosis and is mediated by gasdermin proteins. Pyroptosis is caused by multiple pathways that eventually form pores in the cell membrane, facilitating the release of inflammatory substances and causing the cell to rupture and die. Pyroptosis occurs in neurons, glial cells, and endothelial cells after ICH. Furthermore, pyroptosis causes cell death and releases inflammatory factors such as interleukin (IL)-1β and IL-18, leading to a secondary immune-inflammatory response and further brain damage. The NOD-like receptor protein 3 (NLRP3)/caspase-1/gasdermin D (GSDMD) pathway plays the most critical role in pyroptosis after ICH. Pyroptosis can be inhibited by directly targeting NLRP3 or its upstream molecules, or directly interfering with caspase-1 expression and GSDMD formation, thus significantly improving the prognosis of ICH. The present review discusses key pathological pathways and regulatory mechanisms of pyroptosis after ICH and suggests possible intervention strategies to mitigate pyroptosis and brain dysfunction after ICH.
Collapse
Affiliation(s)
- Dengpan Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chi-Tai Yeh
- Department of Medical Research and Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| | - Fuyou Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| |
Collapse
|
33
|
Tian C, Yang J, Xie N, Tang Y, Zhou H, Hu ZY, Ouyang Q. The prognosis and risk factors for capecitabine maintenance treatment in metastatic breast cancer: a retrospective comparative cohort study. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:924. [PMID: 36172110 PMCID: PMC9511179 DOI: 10.21037/atm-22-3828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/31/2022] [Indexed: 11/06/2022]
Abstract
Background Maintenance treatment following efficient chemotherapy can improve the treatment outcomes of patients with metastatic breast cancer (MBC). However, there are no studies for identifying the prognostic factors for patients who could benefit from capecitabine maintenance. Therefore, this study aimed to investigate the prognosis and risk factors of capecitabine maintenance therapy and analysed the circulating tumour DNA (ctDNA) markers that may be related to the treatment response. Methods This study recruited 482 consecutive patients with MBC who achieved clinical benefit from capecitabine-based chemotherapy from 2011 to 2019. A total of 256 patients received subsequent capecitabine maintenance therapy. The baseline clinical factors included age at diagnosis, menopause, neoadjuvant therapy, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) status and subtypes, prior treatment lines, and prior capecitabine-based treatment response. Treatment outcome (progression-free survival, PFS) was assessed by imaging tools according to RSCIST 1.1 standard during the first two treatment cycles and every 3 weeks thereafter. Univariate and multivariate Cox proportional hazards models were used to analysethe association between capecitabine maintenance treatment and prognosis. Results The median PFS of patients receiving capecitabine maintenance treatment was 21.7 months [95% confidence interval (CI): 15.1-36.3 months]. Capecitabine maintenance showed similar effects as endocrine maintenance or anti-HER2 therapy in hormone receptor (HR)-positive or HER2-positive patients, with adjusted HR of 1.17 (95% CI: 0.81-1.71, P=0.40). In patients with triple-negative breast cancer (TNBC), capecitabine maintenance showed a marginal benefit in PFS. Compared to late-line (≥2) capecitabine maintenance, first-line capecitabine maintenance significantly prolonged median PFS. Compared to other HR/HER2 subtypes, patients with HR-positive and HER2-positive subtypes significantly benefited from capecitabine maintenance treatment. Analysis of ctDNA revealed that among patients receiving capecitabine maintenance, TP53 aberrations were concentrated in patients with short PFS. Conclusions Capecitabine maintenance treatment is associated with longer PFS in patients with MBC, especially those receiving first-line capecitabine-based chemotherapy and those with HR positivity/HER2 positivity. TP53 aberrations may be responsible for the poor response to capecitabine maintenance treatment.
Collapse
Affiliation(s)
- Can Tian
- Medical Department of Breast Cancer, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Jianbo Yang
- The Immunotherapy Research Laboratory, Department of Otolaryngology, University of Minnesota, Minneapolis, MN, USA.,The Cancer Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ning Xie
- Medical Department of Breast Cancer, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Yu Tang
- Medical Department of Breast Cancer, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Haoyu Zhou
- College of Information and Intelligence, Hunan Agricultural University, Changsha, China
| | - Zhe-Yu Hu
- Medical Department of Breast Cancer, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Quchang Ouyang
- Medical Department of Breast Cancer, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| |
Collapse
|
34
|
Sun C, Han Y, Zhang R, Liu S, Wang J, Zhang Y, Chen X, Jiang C, Wang J, Fan X, Wang J. Regulated necrosis in COVID-19: A double-edged sword. Front Immunol 2022; 13:917141. [PMID: 36090995 PMCID: PMC9452688 DOI: 10.3389/fimmu.2022.917141] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
COVID-19 caused by SARS-CoV-2 can cause various systemic diseases such as acute pneumonia with cytokine storm. Constituted of necroptosis, pyroptosis, and ferroptosis, regulated necrosis constitutes the cell death patterns under the low apoptosis condition commonly observed in COVID-19. Regulated necrosis is involved in the release of cytokines like TNF-α, IL-1 β, and IL-6 and cell contents such as alarmins, PAMPs, and DAMPs, leading to more severe inflammation. Uncontrolled regulated necrosis may explain the poor prognosis and cytokine storm observed in COVID-19. In this review, the pathophysiology and mechanism of regulated necrosis with the double-edged sword effect in COVID-19 are thoroughly discussed in detail. Furthermore, this review also focuses on the biomarkers and potential therapeutic targets of the regulated necrosis pathway in COVID-19, providing practical guidance to judge the severity, prognosis, and clinical treatment of COVID-19 and guiding the development of clinical anti-SARS-CoV-2 drugs.
Collapse
Affiliation(s)
- Chen Sun
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunze Han
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruoyu Zhang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Simon Liu
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, MD, United States
| | - Jing Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuqing Zhang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chao Jiang
- Department of Neurology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Jian Wang, ; Junmin Wang, ; Xiaochong Fan,
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Jian Wang, ; Junmin Wang, ; Xiaochong Fan,
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Jian Wang, ; Junmin Wang, ; Xiaochong Fan,
| |
Collapse
|
35
|
Pang Q, Zheng L, Ren Z, Xu H, Guo H, Shan W, Liu R, Gu Z, Wang T. Mechanism of Ferroptosis and Its Relationships with Other Types of Programmed Cell Death: Insights for Potential Therapeutic Benefits in Traumatic Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1274550. [PMID: 36062196 PMCID: PMC9433211 DOI: 10.1155/2022/1274550] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/19/2022] [Accepted: 08/13/2022] [Indexed: 12/05/2022]
Abstract
Traumatic brain injury (TBI) is a serious health issue with a high incidence, high morbidity, and high mortality that poses a large burden on society. Further understanding of the pathophysiology and cell death models induced by TBI may support targeted therapies for TBI patients. Ferroptosis, a model of programmed cell death first defined in 2012, is characterized by iron dyshomeostasis, lipid peroxidation, and glutathione (GSH) depletion. Ferroptosis is distinct from apoptosis, autophagy, pyroptosis, and necroptosis and has been shown to play a role in secondary brain injury and worsen long-term outcomes after TBI. This review systematically describes (1) the regulatory pathways of ferroptosis after TBI, (2) the neurobiological links between ferroptosis and other cell death models, and (3) potential therapies targeting ferroptosis for TBI patients.
Collapse
Affiliation(s)
- Qiuyu Pang
- Department of Forensic Science, Suzhou Medicine College of Soochow University, Suzhou 215123, China
| | - Lexin Zheng
- Department of Forensic Science, Suzhou Medicine College of Soochow University, Suzhou 215123, China
| | - Zhiyang Ren
- Department of Forensic Science, Suzhou Medicine College of Soochow University, Suzhou 215123, China
| | - Heng Xu
- Department of Forensic Science, Suzhou Medicine College of Soochow University, Suzhou 215123, China
| | - Hanmu Guo
- Department of Forensic Science, Suzhou Medicine College of Soochow University, Suzhou 215123, China
| | - Wenqi Shan
- Department of Forensic Science, Suzhou Medicine College of Soochow University, Suzhou 215123, China
| | - Rong Liu
- Department of Forensic Science, Suzhou Medicine College of Soochow University, Suzhou 215123, China
| | - Zhiya Gu
- Department of Forensic Science, Suzhou Medicine College of Soochow University, Suzhou 215123, China
| | - Tao Wang
- Department of Forensic Science, Suzhou Medicine College of Soochow University, Suzhou 215123, China
| |
Collapse
|
36
|
Abi-Ghanem C, Jonnalagadda D, Chun J, Kihara Y, Ranscht B. CAQK, a peptide associating with extracellular matrix components targets sites of demyelinating injuries. Front Cell Neurosci 2022; 16:908401. [PMID: 36072569 PMCID: PMC9441496 DOI: 10.3389/fncel.2022.908401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/01/2022] [Indexed: 11/20/2022] Open
Abstract
The destruction of the myelin sheath that encircles axons leads to impairments of nerve conduction and neuronal dysfunctions. A major demyelinating disorder is multiple sclerosis (MS), a progressively disabling disease in which immune cells attack the myelin. To date, there are no therapies to target selectively myelin lesions, repair the myelin or stop MS progression. Small peptides recognizing epitopes selectively exposed at sites of injury show promise for targeting therapeutics in various pathologies. Here we show the selective homing of the four amino acid peptide, cysteine-alanine-lysine glutamine (CAQK), to sites of demyelinating injuries in three different mouse models. Homing was assessed by administering fluorescein amine (FAM)-labeled peptides into the bloodstream of mice and analyzing sites of demyelination in comparison with healthy brain or spinal cord tissue. FAM-CAQK selectively targeted demyelinating areas in all three models and was absent from healthy tissue. At lesion sites, the peptide was primarily associated with the fibrous extracellular matrix (ECM) deposited in interstitial spaces proximal to reactive astrocytes. Association of FAM-CAQK was detected with tenascin-C although tenascin depositions made up only a minor portion of the examined lesion sites. In mice on a 6-week cuprizone diet, FAM-CAQK peptide crossed the nearly intact blood-brain barrier and homed to demyelinating fiber tracts. These results demonstrate the selective targeting of CAQK to demyelinating injuries under multiple conditions and confirm the previously reported association with the ECM. This work sets the stage for further developing CAQK peptide targeting for diagnostic and therapeutic applications aimed at localized myelin repair.
Collapse
|
37
|
Neshasteh-Riz A, Ramezani F, Kookli K, Moghaddas Fazeli S, Motamed A, Nasirinezhad F, Janzadeh A, Hamblin MR, Asadi M. Optimization of the Duration and Dose of Photobiomodulation Therapy (660 nm Laser) for Spinal Cord Injury in Rats. Photobiomodul Photomed Laser Surg 2022; 40:488-498. [DOI: 10.1089/photob.2022.0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ali Neshasteh-Riz
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Keihan Kookli
- International Campus, Iran University of Medical Sciences, Tehran, Iran
- Occupational Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyedalireza Moghaddas Fazeli
- International Campus, Iran University of Medical Sciences, Tehran, Iran
- Occupational Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Motamed
- Islamic Azad University, College of Veterinary Medicine, Karaj, Iran
| | | | - Atousa Janzadeh
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Mohammadreza Asadi
- Department of Medical Physics, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Zhang R, Sun C, Chen X, Han Y, Zang W, Jiang C, Wang J, Wang J. COVID-19-Related Brain Injury: The Potential Role of Ferroptosis. J Inflamm Res 2022; 15:2181-2198. [PMID: 35411172 PMCID: PMC8994634 DOI: 10.2147/jir.s353467] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 pandemic has caused devastating loss of life and a healthcare crisis worldwide. SARS-CoV-2 is the causative pathogen of COVID-19 and is transmitted mainly through the respiratory tract, where the virus infects host cells by binding to the ACE2 receptor. SARS-CoV-2 infection is associated with acute pneumonia, but neuropsychiatric symptoms and different brain injuries are also present. The possible routes by which SARS-CoV-2 invades the brain are unclear, as are the mechanisms underlying brain injuries with the resultant neuropsychiatric symptoms in patients with COVID-19. Ferroptosis is a unique iron-dependent form of non-apoptotic cell death, characterized by lipid peroxidation with high levels of glutathione consumption. Ferroptosis plays a primary role in various acute and chronic brain diseases, but to date, ferroptosis in COVID-19-related brain injuries has not been explored. This review discusses the mechanisms of ferroptosis and recent evidence suggesting a potential pathogenic role for ferroptosis in COVID-19-related brain injury. Furthermore, the possible routes through which SARS-CoV-2 could invade the brain are also discussed. Discoveries in these areas will open possibilities for treatment strategies to prevent or reduce brain-related complications of COVID-19.
Collapse
Affiliation(s)
- Ruoyu Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, People’s Republic of China
| | - Chen Sun
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, People’s Republic of China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, People’s Republic of China
| | - Yunze Han
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, People’s Republic of China
| | - Weidong Zang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, People’s Republic of China
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, People’s Republic of China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, People’s Republic of China
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, People’s Republic of China
- Correspondence: Jian Wang; Junmin Wang, Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China, Email ;
| |
Collapse
|
39
|
Deficiency in Retinal TGFβ Signaling Aggravates Neurodegeneration by Modulating Pro-Apoptotic and MAP Kinase Pathways. Int J Mol Sci 2022; 23:ijms23052626. [PMID: 35269767 PMCID: PMC8910086 DOI: 10.3390/ijms23052626] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Transforming growth factor β (TGFβ) signaling has manifold functions such as regulation of cell growth, differentiation, migration, and apoptosis. Moreover, there is increasing evidence that it also acts in a neuroprotective manner. We recently showed that TGFβ receptor type 2 (Tgfbr2) is upregulated in retinal neurons and Müller cells during retinal degeneration. In this study we investigated if this upregulation of TGFβ signaling would have functional consequences in protecting retinal neurons. To this end, we analyzed the impact of TGFβ signaling on photoreceptor viability using mice with cell type-specific deletion of Tgfbr2 in retinal neurons and Müller cells (Tgfbr2ΔOC) in combination with a genetic model of photoreceptor degeneration (VPP). We examined retinal morphology and the degree of photoreceptor degeneration, as well as alterations of the retinal transcriptome. In summary, retinal morphology was not altered due to TGFβ signaling deficiency. In contrast, VPP-induced photoreceptor degeneration was drastically exacerbated in double mutant mice (Tgfbr2ΔOC; VPP) by induction of pro-apoptotic genes and dysregulation of the MAP kinase pathway. Therefore, TGFβ signaling in retinal neurons and Müller cells exhibits a neuroprotective effect and might pose promising therapeutic options to attenuate photoreceptor degeneration in humans.
Collapse
|
40
|
Du M, Wu C, Yu R, Cheng Y, Tang Z, Wu B, Fu J, Tan W, Zhou Q, Zhu Z, Balawi E, Huang X, Ma J, Liao ZB. A novel circular RNA, circIgfbp2, links neural plasticity and anxiety through targeting mitochondrial dysfunction and oxidative stress-induced synapse dysfunction after traumatic brain injury. Mol Psychiatry 2022; 27:4575-4589. [PMID: 35918398 PMCID: PMC9734054 DOI: 10.1038/s41380-022-01711-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/14/2022] [Accepted: 07/14/2022] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) can lead to different neurological and psychiatric disorders. Circular RNAs (circRNAs) are highly expressed in the nervous system and enriched in synapses; yet, the underlying role and mechanisms of circRNAs in neurological impairment and dysfunction are still not fully understood. In this study, we investigated the expression of circRNAs and their relation with neurological dysfunction after TBI. RNA-Seq was used to detect differentially expressed circRNAs in injured brain tissue, revealing that circIgfbp2 was significantly increased. Up-regulated hsa_circ_0058195, which was highly homologous to circIgfbp2, was further confirmed in the cerebral cortex specimens and serum samples of patients after TBI. Moreover, correlation analysis showed a positive correlation between hsa_circ_0058195 levels and the Self-Rating Anxiety Scale scores in these subjects. Furthermore, knockdown of circIgfbp2 in mice relieved anxiety-like behaviors and sleep disturbances induced by TBI. Knockdown of circIgfbp2 in H2O2 treated HT22 cells alleviated mitochondrial dysfunction, while its overexpression reversed the process. Mechanistically, we discovered that circIgfbp2 targets miR-370-3p to regulate BACH1, and down-regulating BACH1 alleviated mitochondrial dysfunction and oxidative stress-induced synapse dysfunction. In conclusion, inhibition of circIgfbp2 alleviated mitochondrial dysfunction and oxidative stress-induced synapse dysfunction after TBI through the miR-370-3p/BACH1/HO-1 axis. Thus, circIgfbp2 might be a novel therapeutic target for anxiety and sleep disorders after TBI.
Collapse
Affiliation(s)
- Mengran Du
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Chenrui Wu
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Renqiang Yu
- grid.452206.70000 0004 1758 417XDepartment of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Yuqi Cheng
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Zhaohua Tang
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Biying Wu
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Jiayuanyuan Fu
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Weilin Tan
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Qiang Zhou
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Ziyu Zhu
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Ehab Balawi
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Xuekang Huang
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Jun Ma
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Z. B. Liao
- grid.452206.70000 0004 1758 417XDepartment of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| |
Collapse
|
41
|
Huang Y, Zhao M, Chen X, Zhang R, Le A, Hong M, Zhang Y, Jia L, Zang W, Jiang C, Wang J, Fan X, Wang J. Tryptophan Metabolism in Central Nervous System Diseases: Pathophysiology and Potential Therapeutic Strategies. Aging Dis 2022; 14:858-878. [PMID: 37191427 DOI: 10.14336/ad.2022.0916] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
The metabolism of L-tryptophan (TRP) regulates homeostasis, immunity, and neuronal function. Altered TRP metabolism has been implicated in the pathophysiology of various diseases of the central nervous system. TRP is metabolized through two main pathways, the kynurenine pathway and the methoxyindole pathway. First, TRP is metabolized to kynurenine, then kynurenic acid, quinolinic acid, anthranilic acid, 3-hydroxykynurenine, and finally 3-hydroxyanthranilic acid along the kynurenine pathway. Second, TRP is metabolized to serotonin and melatonin along the methoxyindole pathway. In this review, we summarize the biological properties of key metabolites and their pathogenic functions in 12 disorders of the central nervous system: schizophrenia, bipolar disorder, major depressive disorder, spinal cord injury, traumatic brain injury, ischemic stroke, intracerebral hemorrhage, multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease. Furthermore, we summarize preclinical and clinical studies, mainly since 2015, that investigated the metabolic pathway of TRP, focusing on changes in biomarkers of these neurologic disorders, their pathogenic implications, and potential therapeutic strategies targeting this metabolic pathway. This critical, comprehensive, and up-to-date review helps identify promising directions for future preclinical, clinical, and translational research on neuropsychiatric disorders.
Collapse
|