1
|
Zhao Y, Shen W, Zhang M, Guo M, Dou Y, Han S, Yu J, Cui M, Zhao Y. DDAH-1 maintains endoplasmic reticulum-mitochondria contacts and protects dopaminergic neurons in Parkinson's disease. Cell Death Dis 2024; 15:399. [PMID: 38849335 PMCID: PMC11161642 DOI: 10.1038/s41419-024-06772-w] [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: 11/27/2023] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024]
Abstract
The loss of dopaminergic neurons in the substantia nigra is a hallmark of pathology in Parkinson's disease (PD). Dimethylarginine dimethylaminohydrolase-1 (DDAH-1) is the critical enzyme responsible for the degradation of asymmetric dimethylarginine (ADMA) which inhibits nitric oxide (NO) synthase and has been implicated in neurodegeneration. Mitochondrial dysfunction, particularly in the mitochondria-associated endoplasmic reticulum membrane (MAM), plays a critical role in this process, although the specific molecular target has not yet been determined. This study aims to examine the involvement of DDAH-1 in the nigrostriatal dopaminergic pathway and PD pathogenesis. The distribution of DDAH-1 in the brain and its colocalization with dopaminergic neurons were observed. The loss of dopaminergic neurons and aggravated locomotor disability after rotenone (ROT) injection were showed in the DDAH-1 knockout rat. L-arginine (ARG) and NO donors were employed to elucidate the role of NO respectively. In vitro, we investigated the effects of DDAH-1 knockdown or overexpression on cell viability and mitochondrial functions, as well as modulation of ADMA/NO levels using ADMA or ARG. MAM formation was assessed by the Mitofusin2 oligomerization and the mitochondrial ubiquitin ligase (MITOL) phosphorylation. We found that DDAH-1 downregulation resulted in enhanced cell death and mitochondrial dysfunctions, accompanied by elevated ADMA and reduced NO levels. However, the recovered NO level after the ARG supplement failed to exhibit a protective effect on mitochondrial functions and partially restored cell viability. DDAH-1 overexpression prevented ROT toxicity, while ADMA treatment attenuated these protective effects. The declines of MAM formation in ROT-treated cells were exacerbated by DDAH-1 downregulation via reduced MITOL phosphorylation, which was reversed by DDAH-1 overexpression. Together, the abundant expression of DDAH-1 in nigral dopaminergic neurons may exert neuroprotective effects by maintaining MAM formation and mitochondrial function probably via ADMA, indicating the therapeutic potential of targeting DDAH-1 for PD.
Collapse
Affiliation(s)
- Yichen Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weiwei Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Minjie Zhang
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Guo
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunxiao Dou
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Sida Han
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jintai Yu
- Department of Neurology, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Mei Cui
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yanxin Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Gao Q, Ni P, Wang Y, Huo P, Zhang X, Wang S, Xiao F, Li Y, Feng W, Yuan J, Zhang T, Li Q, Fan B, Kan Y, Li Z, Qi Y, Xing J, Yang Z, Cheng H, Gao X, Feng X, Xue M, Liu Y, Luo Y, Lu Z, Zhao Y. DDAH1 promotes neurogenesis and neural repair in cerebral ischemia. Acta Pharm Sin B 2024; 14:2097-2118. [PMID: 38799640 PMCID: PMC11119513 DOI: 10.1016/j.apsb.2024.02.001] [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: 09/19/2023] [Revised: 12/21/2023] [Accepted: 01/22/2024] [Indexed: 05/29/2024] Open
Abstract
Choline acetyltransferase (ChAT)-positive neurons in neural stem cell (NSC) niches can evoke adult neurogenesis (AN) and restore impaired brain function after injury, such as acute ischemic stroke (AIS). However, the relevant mechanism by which ChAT+ neurons develop in NSC niches is poorly understood. Our RNA-seq analysis revealed that dimethylarginine dimethylaminohydrolase 1 (DDAH1), a hydrolase for asymmetric NG,NG-dimethylarginine (ADMA), regulated genes responsible for the synthesis and transportation of acetylcholine (ACh) (Chat, Slc5a7 and Slc18a3) after stroke insult. The dual-luciferase reporter assay further suggested that DDAH1 controlled the activity of ChAT, possibly through hypoxia-inducible factor 1α (HIF-1α). KC7F2, an inhibitor of HIF-1α, abolished DDAH1-induced ChAT expression and suppressed neurogenesis. As expected, DDAH1 was clinically elevated in the blood of AIS patients and was positively correlated with AIS severity. By comparing the results among Ddah1 general knockout (KO) mice, transgenic (TG) mice and wild-type (WT) mice, we discovered that DDAH1 upregulated the proliferation and neural differentiation of NSCs in the subgranular zone (SGZ) under ischemic insult. As a result, DDAH1 may promote cognitive and motor function recovery against stroke impairment, while these neuroprotective effects are dramatically suppressed by NSC conditional knockout of Ddah1 in mice.
Collapse
Affiliation(s)
- Qiming Gao
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Pinfei Ni
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yilin Wang
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Peiyun Huo
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaojie Zhang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Sihan Wang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Fuyao Xiao
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yixuan Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wei Feng
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juntao Yuan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Teng Zhang
- Department of Laboratory Animal, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Qiang Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Boyu Fan
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yuhao Kan
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Zhirui Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yimiao Qi
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Junfei Xing
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Zhenghong Yang
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Haixiao Cheng
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xinran Gao
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaoyan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Ming Xue
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yang Liu
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yumin Luo
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
3
|
Bi M, Li D, Zhang J. Role of curcumin in ischemia and reperfusion injury. Front Pharmacol 2023; 14:1057144. [PMID: 37021057 PMCID: PMC10067738 DOI: 10.3389/fphar.2023.1057144] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 02/23/2023] [Indexed: 03/22/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) is an inevitable pathological process after organic transplantations. Although traditional treatments restore the blood supply of ischemic organs, the damage caused by IRI is always ignored. Therefore, the ideal and effective therapeutic strategy to mitigate IRI is warrented. Curcumin is a type of polyphenols, processing such properties as anti-oxidative stress, anti-inflammation and anti-apoptosis. However, although many researches have been confirmed that curcumin can exert great effects on the mitigation of IRI, there are still some controversies about its underlying mechanisms among these researches. Thus, this review is to summarize the protective role of curcumin against IRI as well as the controversies of current researches, so as to clarify its underlying mechanisms clearly and provide clinicians a novel idea of the therapy for IRI.
Collapse
Affiliation(s)
- Minglei Bi
- Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Danyi Li
- Department of Ophthalmology, Jiading District Central Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jin Zhang
- Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- *Correspondence: Jin Zhang,
| |
Collapse
|
4
|
DDAH1/ADMA Regulates Adiponectin Resistance in Cerebral Ischemia via the ROS/FOXO1/APR1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2350857. [PMID: 35509834 PMCID: PMC9060971 DOI: 10.1155/2022/2350857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/14/2022] [Accepted: 03/30/2022] [Indexed: 11/27/2022]
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) protects against cerebral ischemia injury via regulating the level of asymmetric dimethylarginine (ADMA). This study is aimed at exploring the effect of adiponectin resistance on ADMA-induced neuronal loss in ischemic stroke (IS) and the underlying mechanism. DDAH1 knockout (DDAH1−/−) and wild-type (DDAH1+/+) rats underwent middle cerebral artery occlusion/reperfusion (MCAO/R). Plasma and brain adiponectin levels and the expressions of adiponectin receptor 1 (APR1), adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1), adenosine monophosphate-activated protein kinase (AMPK), and phosphorylated AMPK were determined after 24 h, 3 days, and 7 days. Neurological behavior, infarct volume, and adiponectin signaling were evaluated using adiponectin peptide or AdipoRon. The levels of reactive oxygen species (ROS) and Forkhead box O1 (FOXO1) (a transcription factor for APR1) were also assessed. An oxygen-glucose deprivation/reoxygenation (OGD/R) model was established in primary neurons. DDAH1 was overexpressed in neurons, after which FOXO1 expression, ROS production, adiponectin resistance, and cell viability were detected. DDAH1−/− rats showed no significant difference in adiponectin level in either plasma or brain after MCAO/R in DDAH1+/+ rats, but downregulated APR1 expression and suppressed adiponectin signaling were observed. AdipoRon, but not adiponectin peptide, attenuated the neurological deficits and adiponectin resistance in DDAH1−/− rats. ROS accumulation and phosphorylated FOXO1 expression also increased with DDAH1 depletion. Following DDAH1 overexpression, decreased cell viability and inhibited adiponectin signaling induced by OGD/R were alleviated in primary neurons, accompanied by reduced ROS production and phosphorylated FOXO1 expression. Our study elucidated that in IS, DDAH1 protected against adiponectin resistance in IS via the ROS/FOXO1/APR1 pathway.
Collapse
|
5
|
Roles of Nitric Oxide in Brain Ischemia and Reperfusion. Int J Mol Sci 2022; 23:ijms23084243. [PMID: 35457061 PMCID: PMC9028809 DOI: 10.3390/ijms23084243] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 01/27/2023] Open
Abstract
Brain ischemia and reperfusion (I/R) is one of the most severe clinical manifestations of ischemic stroke, placing a significant burden on both individuals and society. The only FDA-approved clinical treatment for ischemic stroke is tissue plasminogen activator (t-PA), which rapidly restores cerebral blood flow but can have severe side effects. The complex pathological process of brain I/R has been well-established in the past few years, including energy metabolism disorders, cellular acidosis, doubling of the synthesis or release of excitotoxic amino acids, intracellular calcium homeostasis, free radical production, and activation of apoptotic genes. Recently, accumulating evidence has shown that NO may be strongly related to brain I/R and involved in complex pathological processes. This review focuses on the role of endogenous NO in pathological processes in brain I/R, including neuronal cell death and blood brain barrier disruption, to explore how NO impacts specific signaling cascades and contributes to brain I/R injury. Moreover, NO can rapidly react with superoxide to produce peroxynitrite, which may also mediate brain I/R injury, which is discussed here. Finally, we reveal several therapeutic approaches strongly associated with NO and discuss their potential as a clinical treatment for ischemic stroke.
Collapse
|
6
|
Wan Y, Huang L, Liu Y, Ji W, Li C, Ge RL. Preconditioning With Intermittent Hypobaric Hypoxia Attenuates Stroke Damage and Modulates Endocytosis in Residual Neurons. Front Neurol 2022; 12:750908. [PMID: 34975719 PMCID: PMC8715922 DOI: 10.3389/fneur.2021.750908] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/26/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Moderate hypobaric hypoxia induces cerebral ischemic tolerance. We investigated the optimal method for applying hypobaric hypoxia preconditioning at 5,000 m to ischemic brain tissue and combined it with proteomics to determine the mechanisms underlying this effect. Methods: Male SD rats were randomly grouped as S (sham, n = 20), M (middle cerebral artery occlusion [MCAO], n = 28), H2M (intermittent hypobaric hypoxia preconditioned MCAO group, 2 h/day, 10 days, n = 20), H6M (intermittent hypobaric hypoxia preconditioned MCAO group, 6 h/day, 10 days, n = 28), and HpM (persistent hypobaric hypoxia preconditioned MCAO group, 10 days, n = 28). The permanent MCAO model was established based on the Zea Longa method. Infarction was assessed with the modified neurological severity score (mNSS) and 2,3,5-triphenyl tetrazolium chloride staining. The total protein expression of the neuron-specific nuclear protein (NeuN), cysteinyl aspartate specific proteinase 3 (caspase-3), cleaved-caspase-3, and interleukin 6 (IL-6) was determined using western blotting. We assessed the peri-infarct cortex's ultrastructural changes. A label-free proteomic study and western blot verification were performed on the most effective preconditioned group. Results: The H6M group showed a lower infarct volume (p = 0.0005), lower mNSS score (p = 0.0009) than the M group. The H2M showed a lower level of IL-6 (p = 0.0213) than the M group. The caspase-3 level decreased in the H2M (p = 0.0002), H6M (p = 0.0025), and HpM groups (p = 0.0054) compared with that in the M group. Cleaved-caspase-3 expression decreased in the H2M (p = 0.0011), H6M (p < 0.0001), and HpM groups (p < 0.0001) compared with that in the M group. The neurons' ultrastructure and the blood-brain barrier in the peri-infarct tissue improved in the H2M and H6M groups. Immunofluorescence revealed increased NeuN-positive cells in the peri-infarct tissue in the H6M group (p = 0.0003, H6M vs. M). Protein expression of Chmp1a, Arpc5, and Hspa2 factors related to endocytosis were upregulated in the H6M compared with those of the M group (p < 0.05 for all) on western blot verification of label-free proteomics. Conclusions: Intermittent hypobaric hypoxia preconditioning exerts a neuroprotective effect in a rat stroke model. Persistent hypobaric hypoxia stimulation exhibited no significant neuroprotective effect. Intermittent hypoxic preconditioning for 6 h/day for 10 days upregulates key proteins in clathrin-dependent endocytosis of neurons in the cortex.
Collapse
Affiliation(s)
- Yaqi Wan
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Research Center for High Altitude Medicine, Qinghai University, Xining, China.,Qinghai Provincial People's Hospital, Xining, China
| | - Lu Huang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanmin Liu
- Qinghai Provincial People's Hospital, Xining, China
| | - Weizhong Ji
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Research Center for High Altitude Medicine, Qinghai University, Xining, China.,Qinghai Provincial People's Hospital, Xining, China
| | - Changxing Li
- Department of Basic Medicine, Qinghai University, Xining, China
| | - Ri-Li Ge
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Research Center for High Altitude Medicine, Qinghai University, Xining, China
| |
Collapse
|
7
|
Uyeda A, Quan L, Kato Y, Muramatsu N, Tanabe S, Sakai K, Ichinohe N, Kawahara Y, Suzuki T, Muramatsu R. Dimethylarginine dimethylaminohydrolase 1 as a novel regulator of oligodendrocyte differentiation in the central nervous system remyelination. Glia 2021; 69:2591-2604. [PMID: 34270117 DOI: 10.1002/glia.24060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/12/2022]
Abstract
Remyelination is a regenerative process that restores the lost neurological function and partially depends on oligodendrocyte differentiation. Differentiation of oligodendrocytes spontaneously occurs after demyelination, depending on the cell intrinsic mechanisms. By combining a loss-of-function genomic screen with a web-resource-based candidate gene identification approach, we identified that dimethylarginine dimethylaminohydrolase 1 (DDAH1) is a novel regulator of oligodendrocyte differentiation. Silencing DDAH1 in oligodendrocytes prevented the expression of myelin basic protein in mouse oligodendrocyte culture with the change in expression of genes annotated with oligodendrocyte development. DDAH1 inhibition attenuated spontaneous remyelination in a cuprizone-induced demyelinated mouse model. Conversely, increased DDAH1 expression enhanced remyelination capacity in experimental autoimmune encephalomyelitis. These results provide a novel therapeutic option for demyelinating diseases by modulating DDAH1 activity.
Collapse
Affiliation(s)
- Akiko Uyeda
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Lili Quan
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yuki Kato
- Department of RNA Biology and Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Nagaaki Muramatsu
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Department of Medical and Life Science, Graduate School of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Shogo Tanabe
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Kazuhisa Sakai
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Noritaka Ichinohe
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yukio Kawahara
- Department of RNA Biology and Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tatsunori Suzuki
- Department of Medical and Life Science, Graduate School of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Rieko Muramatsu
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| |
Collapse
|
8
|
Zhao Y, Ma X, Zhou Y, Xie J, Liu X, Zhao Y. DDAH-1, via regulation of ADMA levels, protects against ischemia-induced blood-brain barrier leakage. J Transl Med 2021; 101:808-823. [PMID: 33574439 DOI: 10.1038/s41374-021-00541-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 01/01/2023] Open
Abstract
Dimethylarginine dimethylamino hydrolase-1 (DDAH-1) is an important regulator of nitric oxide (NO) metabolism that has been implicated in the pathogenesis of cardiovascular diseases. Nevertheless, its role in cerebral ischemia still needs to be elucidated. Herein, we examined the expression of DDAH-1 in the brain of rat by double-label immunofluorescence staining. DDAH-1 knock-out (DDAH-1-/-) and wild-type rats underwent middle cerebral artery occlusion/reperfusion (MCAO/R). After 24 h, neurological scores, TTC staining and TUNEL assay were used to evaluate neurological damages. 3 and 7-days infarct outcomes were also shown. Blood-brain-barrier (BBB) permeability was examined via Evans blue extravasation and tight junction (TJ) proteins expression and mRNA levels by western blot and RT-qPCR. The levels of plasma asymmetric dimethylarginine (ADMA), NO and ADMA in brain tissue were also assessed. In addition, supplementation of L-arginine to DDAH-1-/- rats was used to explore its role in regulating NO. DDAH-1 was abundantly distributed in cerebral cortex and basal nuclei, and mainly expressed in neurons and endothelial cells. DDAH-1-/- rats showed aggravated neurological damage and BBB disruption, including decrease of TJ proteins expression but indistinguishable mRNA levels after MCAO/R. DDAH-1 depletion and neurological damages were accompanied with increased ADMA levels and decreased NO concentrations. The supplementation with L-arginine partly restored the neurological damages and BBB disruption. To sum up, DDAH-1 revealed to have a protective role in ischemia stroke (IS) and IS-induced leakage of BBB via decreasing ADMA level and possibly via preventing TJ proteins degradation.
Collapse
Affiliation(s)
- Yichen Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301 Middle Yanchang Road, Shanghai, 200072, PR China
| | - Xiaoye Ma
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301 Middle Yanchang Road, Shanghai, 200072, PR China
| | - Yuchen Zhou
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301 Middle Yanchang Road, Shanghai, 200072, PR China
| | - Junchao Xie
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301 Middle Yanchang Road, Shanghai, 200072, PR China
| | - Xueyuan Liu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301 Middle Yanchang Road, Shanghai, 200072, PR China.
| | - Yanxin Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301 Middle Yanchang Road, Shanghai, 200072, PR China.
| |
Collapse
|
9
|
Wang L, Ren C, Li Y, Gao C, Li N, Li H, Wu D, He X, Xia C, Ji X. Remote ischemic conditioning enhances oxygen supply to ischemic brain tissue in a mouse model of stroke: Role of elevated 2,3-biphosphoglycerate in erythrocytes. J Cereb Blood Flow Metab 2021; 41:1277-1290. [PMID: 32933360 PMCID: PMC8142126 DOI: 10.1177/0271678x20952264] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oxygen supply for ischemic brain tissue during stroke is critical to neuroprotection. Remote ischemic conditioning (RIC) treatment is effective for stroke. However, it is not known whether RIC can improve brain tissue oxygen supply. In current study, we employed a mouse model of stroke created by middle cerebral artery occlusion (MCAO) to investigate the effect of RIC on oxygen supply to the ischemic brain tissue using a hypoxyprobe system. Erythrocyte oxygen-carrying capacity and tissue oxygen exchange were assessed by measuring oxygenated hemoglobin and oxygen dissociation curve. We found that RIC significantly mitigated hypoxic signals and decreased neural cell death, thereby preserving neurological functions. The tissue oxygen exchange was markedly enhanced, along with the elevated hemoglobin P50 and right-shifted oxygen dissociation curve. Intriguingly, RIC markedly elevated 2,3-biphosphoglycerate (2,3-BPG) levels in erythrocyte, and the erythrocyte 2,3-BPG levels were highly negatively correlated with the hypoxia in the ischemic brain tissue. Further, adoptive transfusion of 2,3-BPG-rich erythrocytes prepared from RIC-treated mice significantly enhanced the oxygen supply to the ischemic tissue in MCAO mouse model. Collectively, RIC protects against ischemic stroke through improving oxygen supply to the ischemic brain tissue where the enhanced tissue oxygen delivery and exchange by RIC-induced 2,3-BPG-rich erythrocytes may play a role.
Collapse
Affiliation(s)
- Lin Wang
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Yang Li
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chen Gao
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ning Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haiyan Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Di Wu
- Deparment of Neurology, China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoduo He
- Deparment of Neurology, China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Changqing Xia
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Municipal Geriatric Medical Research Center, Beijing, China.,Deparment of Neurology, China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| |
Collapse
|
10
|
Grosse GM, Schwedhelm E, Worthmann H, Choe CU. Arginine Derivatives in Cerebrovascular Diseases: Mechanisms and Clinical Implications. Int J Mol Sci 2020; 21:ijms21051798. [PMID: 32150996 PMCID: PMC7084464 DOI: 10.3390/ijms21051798] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 02/07/2023] Open
Abstract
The amino acid L-arginine serves as substrate for the nitric oxide synthase which is crucial in vascular function and disease. Derivatives of arginine, such as asymmetric (ADMA) and symmetric dimethylarginine (SDMA), are regarded as markers of endothelial dysfunction and have been implicated in vascular disorders. While there is a variety of studies consolidating ADMA as biomarker of cerebrovascular risk, morbidity and mortality, SDMA is currently emerging as an interesting metabolite with distinct characteristics in ischemic stroke. In contrast to dimethylarginines, homoarginine is inversely associated with adverse events and mortality in cerebrovascular diseases and might constitute a modifiable protective risk factor. This review aims to provide an overview of the current evidence for the pathophysiological role of arginine derivatives in cerebrovascular ischemic diseases. We discuss the complex mechanisms of arginine metabolism in health and disease and its potential clinical implications in diverse aspects of ischemic stroke.
Collapse
Affiliation(s)
- Gerrit M. Grosse
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany;
- Correspondence:
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20249 Hamburg, Germany;
- DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, 20249 Hamburg, Germany
| | - Hans Worthmann
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany;
| | - Chi-un Choe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20249 Hamburg, Germany;
| |
Collapse
|