1
|
Liu QY, Cui Y, Li W, Qiu J, Nguyen TN, Chen HS. Effect of remote ischemic preconditioning on cerebral circulation time in severe carotid artery stenosis: Results from the RIC-CCT trial. Cell Rep Med 2024; 5:101796. [PMID: 39471820 PMCID: PMC11604480 DOI: 10.1016/j.xcrm.2024.101796] [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: 07/10/2024] [Revised: 08/21/2024] [Accepted: 09/27/2024] [Indexed: 11/01/2024]
Abstract
In patients with severe internal carotid artery stenosis (sICAS), cerebral circulation time (CCT) is associated with cerebral hyperperfusion syndrome. This study aims to investigate the effect of remote ischemic preconditioning (RIC) on CCT in patients with sICAS. Patients are randomly assigned to the RIC group (RIC twice daily, for 2-4 days before carotid artery stenting [CAS] as an adjunct to standard medical therapy) and the control group. The results show that RIC produces a significant decrease in CCT of the stenosis side (sCCT) from baseline to pre-CAS, and the occurrence of contrast staining on brain computed tomography (CT) is lower in RIC versus control group after CAS. In addition, significant changes in some serum biomarkers suggest that anti-neuroinflammation, anti-oxidative stress, protecting endothelial injury, and improving cerebral autoregulation may be associated with the effect of RIC. These findings provide supporting evidence that RIC can modulate cerebral circulation in patients with sICAS. This study was registered at ClinicalTrials.gov (NCT05451030).
Collapse
Affiliation(s)
- Quan-Ying Liu
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Yu Cui
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Wei Li
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Jing Qiu
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Thanh N Nguyen
- Department of Neurology, Radiology, Boston Medical Center, Boston, MA, USA
| | - Hui-Sheng Chen
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China.
| |
Collapse
|
2
|
Qin L, Tong F, Li S, Ren C. Beyond Pharmacology: The Biological Mechanisms of Remote Ischemic Conditioning in Cerebrovascular Disease. Biomolecules 2024; 14:1408. [PMID: 39595584 PMCID: PMC11592304 DOI: 10.3390/biom14111408] [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: 09/24/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Cerebrovascular diseases (CVDs), comprising predominantly ischemic stroke and chronic cerebral hypoperfusion (CCH), are a significant threat to global health, often leading to disability and mortality. Remote ischemic conditioning (RIC) has emerged as a promising, non-pharmacological strategy to combat CVDs by leveraging the body's innate defense mechanisms. This review delves into the neuroprotective mechanisms of RIC, categorizing its effects during the acute and chronic phases of stroke recovery. It also explores the synergistic potential of RIC when combined with other therapeutic strategies, such as pharmacological treatments and physical exercise. Additionally, this review discusses the pathways through which peripheral transmission can confer central neuroprotection. This review concludes by addressing the challenges regarding and future directions for RIC, emphasizing the need for standardized protocols, biomarker identification, and expanded clinical trials to fully realize its therapeutic potential.
Collapse
Affiliation(s)
| | | | | | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (L.Q.); (F.T.); (S.L.)
| |
Collapse
|
3
|
Liu J, Yang N, Wang X, Wang W. Piracetam reduces oxidative stress and mitochondrial function impairment in an in vitro model of vascular dementia. Exp Brain Res 2024; 242:1841-1850. [PMID: 38842755 DOI: 10.1007/s00221-024-06868-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Vascular dementia (VaD) is the most common cause of dementia in older adults. Due to the lack of effective treatment options, there is an urgent need to find an effective pharmaceutical compound to combat VaD. Piracetam has been reported to improve impaired cognitive function in a variety of conditions in both human and animal models. However, the role and mechanism of Piracetam in VaD remain unclear. Therefore this study aimed to elucidate the effect of Piracetam on a cellular model of VaD in vitro. We found that Piracetam enhanced the growth of OGD-stimulated SH-SY5Y cells. In addition, Piracetam inhibited the oxidative stress of OGD-stimulated SH-SY5Y cells. Further, Piracetam improved mitochondrial function of OGD-stimulated SH-SY5Y cells. Mechanistically, Piracetam inhibited the PI3K/Akt/mTOR pathway in OGD-stimulated SH-SY5Y cells. Collectively, Piracetam improved oxidative stress and mitochondrial dysfunction of OGD-stimulated SH-SY5Y cells through PI3K/Akt/mTOR axis. Hence, Piracetam has the potential to serve as a promising drug of VaD.
Collapse
Affiliation(s)
- Juan Liu
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, No.48 FengHao Road(West), Lianhu District, Xi'an City, 710077, Shaanxi Province, China.
| | - Na Yang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, No.48 FengHao Road(West), Lianhu District, Xi'an City, 710077, Shaanxi Province, China
| | - Xiaomeng Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, No.48 FengHao Road(West), Lianhu District, Xi'an City, 710077, Shaanxi Province, China
| | - Wen Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, No.48 FengHao Road(West), Lianhu District, Xi'an City, 710077, Shaanxi Province, China
| |
Collapse
|
4
|
Blauenfeldt RA, Mortensen JK, Hjort N, Valentin JB, Homburg AM, Modrau B, Sandal BF, Gude MF, Berhndtz AB, Johnsen SP, Hess DC, Simonsen CZ, Andersen G. Effect of Remote Ischemic Conditioning in Ischemic Stroke Subtypes: A Post Hoc Subgroup Analysis From the RESIST Trial. Stroke 2024; 55:874-879. [PMID: 38299363 PMCID: PMC10962424 DOI: 10.1161/strokeaha.123.046144] [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: 12/08/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Remote ischemic conditioning (RIC) is a simple and noninvasive procedure that has proved to be safe and feasible in numerous smaller clinical trials. Mixed results have been found in recent large randomized controlled trials. This is a post hoc subgroup analysis of the RESIST trial (Remote Ischemic Conditioning in Patients With Acute Stroke), investigating the effect of RIC in different acute ischemic stroke etiologies, and whether an effect was modified by treatment adherence. METHODS Eligible patients were adults (aged ≥18 years), independent in activities of daily living, who had prehospital stroke symptoms with a duration of less than 4 hours. They were randomized to RIC or sham. The RIC treatment protocol consisted of 5 cycles with 5 minutes of cuff inflation alternating with 5 minutes with a deflated cuff. Acceptable treatment adherence was defined as when at least 80% of planned RIC cycles were received. The analysis was performed using the entire range (shift analysis) of the modified Rankin Scale (ordinal logistic regression). RESULTS A total of 698 had acute ischemic stroke, 253 (36%) were women, and the median (interquartile range) age was 73 (63-80) years. Median (interquartile range) overall adherence to RIC/sham was 91% (68%-100%). In patients with a stroke due to cerebral small vessel disease, who were adherent to treatment, RIC was associated with improved functional outcome, and the odds ratio for a shift to a lower score on the modified Rankin Scale was 2.54 (1.03-6.25); P=0.042. The association remained significant after adjusting for potential confounders. No significant associations were found with other stroke etiologies, and the overall test for interaction was not statistically significant (χ2, 4.33, P=0.23). CONCLUSIONS In patients with acute ischemic stroke due to cerebral small vessel disease, who maintained good treatment adherence, RIC was associated with improved functional outcomes at 90 days. These results should only serve as a hypothesis-generating for future trials. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03481777.
Collapse
Affiliation(s)
- Rolf Ankerlund Blauenfeldt
- Department of Neurology, Aarhus University Hospital, Denmark (R.A.B., J.K.M., N.H., C.Z.S., G.A.)
- Department of Clinical Medicine, Aarhus University, Denmark (R.A.B., J.K.M., N.H., M.F.G., C.Z.S., G.A.)
| | - Janne Kaergaard Mortensen
- Department of Neurology, Aarhus University Hospital, Denmark (R.A.B., J.K.M., N.H., C.Z.S., G.A.)
- Department of Clinical Medicine, Aarhus University, Denmark (R.A.B., J.K.M., N.H., M.F.G., C.Z.S., G.A.)
| | - Niels Hjort
- Department of Neurology, Aarhus University Hospital, Denmark (R.A.B., J.K.M., N.H., C.Z.S., G.A.)
- Department of Clinical Medicine, Aarhus University, Denmark (R.A.B., J.K.M., N.H., M.F.G., C.Z.S., G.A.)
| | - Jan Brink Valentin
- Department of Clinical Medicine, Danish Center for Health Services Research, Aalborg University, Denmark (J.B.V., S.P.J.)
| | - Anne-Mette Homburg
- Department of Neurology, Research Unit for Neurology, Odense University Hospital, Denmark (A.-M.H.)
| | - Boris Modrau
- Department of Neurology, Aalborg University Hospital, Denmark (B.M.)
| | | | - Martin Faurholdt Gude
- Department of Clinical Medicine, Aarhus University, Denmark (R.A.B., J.K.M., N.H., M.F.G., C.Z.S., G.A.)
- Department of Research and Development, Prehospital Emergency Medical Services, Central Denmark Region, Aarhus, Denmark (M.F.G.)
| | - Anne Brink Berhndtz
- Department of Neurology, Regional Hospital Gødstrup, Denmark (B.F.S., A.B.B.)
| | - Søren Paaske Johnsen
- Department of Clinical Medicine, Danish Center for Health Services Research, Aalborg University, Denmark (J.B.V., S.P.J.)
| | - David C. Hess
- Department of Neurology, Medical College of Georgia, Augusta University, GA (D.C.H.)
| | - Claus Ziegler Simonsen
- Department of Neurology, Aarhus University Hospital, Denmark (R.A.B., J.K.M., N.H., C.Z.S., G.A.)
- Department of Clinical Medicine, Aarhus University, Denmark (R.A.B., J.K.M., N.H., M.F.G., C.Z.S., G.A.)
| | - Grethe Andersen
- Department of Neurology, Aarhus University Hospital, Denmark (R.A.B., J.K.M., N.H., C.Z.S., G.A.)
- Department of Clinical Medicine, Aarhus University, Denmark (R.A.B., J.K.M., N.H., M.F.G., C.Z.S., G.A.)
| |
Collapse
|
5
|
Zhang H, Yang Y, Zhang J, Huang L, Niu Y, Chen H, Liu Q, Wang R. Oligodendrocytes Play a Critical Role in White Matter Damage of Vascular Dementia. Neuroscience 2024; 538:1-10. [PMID: 37913862 DOI: 10.1016/j.neuroscience.2023.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/17/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
With the deepening of population aging, the treatment of cognitive impairment and dementia is facing increasing challenges. Vascular dementia (VaD) is a cognitive dysfunction caused by brain blood flow damage and one of the most common causes of dementia after Alzheimer's disease. White matter damage in patients with chronic ischemic dementia often occurs before cognitive impairment, and its pathological changes include leukoaraiosis, myelin destruction and oligodendrocyte death. The pathophysiology of vascular dementia is complex, involving a variety of neuronal and vascular lesions. The current proposed mechanisms include calcium overload, oxidative stress, nitrative stress and inflammatory damage, which can lead to hypoxia-ischemia and demyelination. Oligodendrocytes are the only myelinating cells in the central nervous system and closely associated with VaD. In this review article, we intend to further discuss the role of oligodendrocytes in white matter and myelin injury in VaD and the development of anti-myelin injury target drugs.
Collapse
Affiliation(s)
- Hexin Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yanrong Yang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Jingjing Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Li Huang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yang Niu
- Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia medical University, Yinchuan 750004, Ningxia, China
| | - Hua Chen
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Qibing Liu
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou 570100, China
| | - Rui Wang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| |
Collapse
|
6
|
Li N, Ren C, Li S, Yu W, Jin K, Ji X. Remote ischemic conditioning alleviates chronic cerebral hypoperfusion-induced cognitive decline and synaptic dysfunction via the miR-218a-5p/SHANK2 pathway. Prog Neurobiol 2023; 230:102514. [PMID: 37574039 DOI: 10.1016/j.pneurobio.2023.102514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/15/2023]
Abstract
Vascular cognitive impairment (VCI) due to chronic cerebral hypoperfusion (CCH), is the second leading cause of dementia. Although synaptic impairment plays a critical role in VCI, its exact mechanism remains unknown. Our previous research revealed that remote ischemic conditioning (RIC) could alleviate cognitive decline resulting from CCH, however, its effects on synaptic impairment remain unclear. In this study, we confirmed that RIC alleviated both cognitive decline and its associated synaptic dysfunction caused by CCH. RNA sequencing revealed that CCH increased in miR-218a-5p expression, which was decreased by RIC. Elevated miR-218a-5p levels limited the benefits of RIC, however, inhibiting miR-218a-5p in hippocampal CA1 neurons rescued synaptic dysfunction. Additionally, we found that SHANK2 is a downstream target of miR-218a-5p, and inhibiting SHANK2 expression reduced the alleviation caused by hypoxic conditioning in synaptic impairment in vitro. In conclusion, our results suggested that RIC alleviated synaptic impairment via the miR-218a-5p/SHANK2 pathway, which could be a potential biomarker or therapeutic target for cognitive impairment caused by CCH.
Collapse
Affiliation(s)
- Ning Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Wantong Yu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Kunlin Jin
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Xuming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorder, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
7
|
Zhang J, Xiao Y, Liu H, Xu L, Guo X, Gao Y, Li M, Xu J, Qi Q, Lv P. Edaravone Dexborneol Alleviates Neuroinflammation by Reducing Neuroglial Cell Proliferation and Suppresses Neuronal Apoptosis/Autophagy in Vascular Dementia Rats. Neurochem Res 2023; 48:3113-3128. [PMID: 37338792 DOI: 10.1007/s11064-023-03973-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/21/2023]
Abstract
More and more evidence shows that the pathological mechanism of vascular dementia (VD) is closely related to oxidative stress injury, cell apoptosis, autophagy, inflammatory response, excitatory amino acid toxicity, synaptic plasticity change, calcium overload, and other processes. Edaravone dexborneol (EDB) is a new type of neuroprotective agent that can improve the neurological damage caused by an ischemic stroke. Previous studies showed that EDB has effects on synergistic antioxidants and induces anti-apoptotic responses. However, it remains unclear whether EDB can affect apoptosis and autophagy by activating the PI3K/Akt/mTOR signaling pathway and its impact on the neuroglial cells. In this study, we established the VD model of rats by bilateral carotid artery occlusion to explore the neuroprotective effect of EDB and its mechanism. Morris Water Maze test was applied to assess the cognitive function of rats. H&E and TUNEL staining were applied to observe the cellular structure of the hippocampus. Immunofluorescence labeling was used to observe the proliferation of astrocytes and microglia. ELISA was applied to examine the levels of TNF-α, IL-1β and IL-6, and RT-PCR was applied to examine their mRNA expression levels. Western blotting was applied to examine apoptosis-related proteins (Bax, Bcl-2, Caspase-3), autophagy-related proteins (Beclin-1, P62, LC3B), PI3K/Akt/mTOR signaling pathway proteins and their phosphorylation levels. The results indicated that EDB ameliorates learning and memory in rats subjected to the VD model, alleviates neuroinflammatory response by reducing the proliferation of the neuroglial cell and inhibits apoptosis and autophagy, which may be mediated by the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jiawei Zhang
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yining Xiao
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Hongna Liu
- Department of Geriatric Gastroenterology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Lili Xu
- Graduate School of Hebei North University, Zhangjiakou, 075000, China
| | - Xing Guo
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, 050051, China
| | - Yaran Gao
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Meixi Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Jing Xu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Qianqian Qi
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, China.
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China.
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Hebei General Hospital, Shijiazhuang, 050051, China.
| |
Collapse
|
8
|
Chen Y, Zhang C. Role of noncoding RNAs in orthodontic tooth movement: new insights into periodontium remodeling. J Transl Med 2023; 21:101. [PMID: 36759852 PMCID: PMC9912641 DOI: 10.1186/s12967-023-03951-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Orthodontic tooth movement (OTM) is biologically based on the spatiotemporal remodeling process in periodontium, the mechanisms of which remain obscure. Noncoding RNAs (ncRNAs), especially microRNAs and long noncoding RNAs, play a pivotal role in maintaining periodontal homeostasis at the transcriptional, post-transcriptional, and epigenetic levels. Under force stimuli, mechanosensitive ncRNAs with altered expression levels transduce mechanical load to modulate intracellular genes. These ncRNAs regulate the biomechanical responses of periodontium in the catabolic, anabolic, and coupling phases throughout OTM. To achieve this, down or upregulated ncRNAs actively participate in cell proliferation, differentiation, autophagy, inflammatory, immune, and neurovascular responses. This review highlights the regulatory mechanism of fine-tuning ncRNAs in periodontium remodeling during OTM, laying the foundation for safe, precise, and personalized orthodontic treatment.
Collapse
Affiliation(s)
- Yuming Chen
- grid.284723.80000 0000 8877 7471Stomatological Hospital, Southern Medical University, Guangzhou, 510280 China
| | - Chao Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
9
|
Li X, Ren C, Li S, Zhao W, Wang P, Ji X. The antihypertensive effect of remote ischemic conditioning in spontaneously hypertensive rats. Front Immunol 2023; 13:1093262. [PMID: 36713422 PMCID: PMC9878686 DOI: 10.3389/fimmu.2022.1093262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023] Open
Abstract
Purpose Limb remote ischemic conditioning (LRIC) may be an effective method to control hypertension. This study investigated whether LRIC decreases blood pressure by regulating the hypertensive inflammatory response in spontaneously hypertensive rats (SHR). Method The SHR and aged-matched Wistar rats with different ages were randomly assigned to the SHR group, SHR+LRIC group, Wistar group, and Wistar + LRIC group. LRIC was conducted by tightening a tourniquet around the upper thigh and releasing it for three cycles daily (10 mins x3 cycles). Blood pressure, the percentage of monocytes and T lymphocytes, and the concentration of pro-inflammatory cytokines in the blood were analyzed. Results The blood pressure of SHR was significantly higher than that of age-matched Wistar rats. LRIC decreased blood pressure in SHR at different ages (4, 8, and 16 weeks old), but had no effect on the blood pressure in Wistar rats. Flow cytometry analysis showed that blood monocytes and CD8 T cells of SHR were higher than those of Wistar rats. LRIC significantly decreased the percentage of monocytes and CD8 T cells in SHR. Consistent with the changes of immune cells, the levels of plasma IL-6 and TNF-α in SHR were also higher. And LRIC attenuated the plasma IL-6 and TNF-α levels in SHR. Conclusion LRIC may decreased the blood pressure via modulation of the inflammatory response in SHR.
Collapse
Affiliation(s)
- Xiaohua Li
- Department of Neurology, Aerospace center Hospital, Beijing, China
- Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Wenbo Zhao
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Peifu Wang
- Department of Neurology, Aerospace center Hospital, Beijing, China
| | - Xunming Ji
- Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Dulaglutide Improves Gliosis and Suppresses Apoptosis/Autophagy Through the PI3K/Akt/mTOR Signaling Pathway in Vascular Dementia Rats. Neurochem Res 2022; 48:1561-1579. [PMID: 36571662 DOI: 10.1007/s11064-022-03853-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/11/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Dulaglutide is a new type of hypoglycemic agent that agonizes glucagon-like peptide-1 receptor (GLP-1RA). It can be concluded from previous studies that a GLP-1RA can reduce apoptosis and regulate autophagy in the nervous system, while related research on dulaglutide in vascular dementia (VD) has not been reported. In our study, the VD rat model was established by bilateral carotid artery occlusion, and the results of the Morris water maze test (MWM) and open-field test showed that the application of dulaglutide could effectively reduce the cognitive decline of VD rats without changing the behavior in the open-field test, which was used to assess an anxiety-like phenotype. We applied HE staining and immunofluorescence labeling to show that dulaglutide treatment significantly alleviated neuronal damage in the hippocampal region of VD rats, and reduced microglial and astrocyte proliferation. Western blot results showed that dulaglutide reduced VD-induced neuronal apoptosis (BCL2/BAX, c-caspase3) and autophagy (P62, LC3B, Beclin-1), and upregulated phosphorylation of PI3K/Akt/mTOR signaling pathway. KEGG pathway analysis of RNA-Sequence results showed that the differentially expressed genes in the dulaglutide treatment group were significantly enriched in the mTOR signaling pathway, and the repressor of mTOR, Deptor, was down-regulated. In conclusion, this study suggested that dulaglutide may alleviate learning and memory impairment and neuron damage in VD rats by attenuating apoptosis, regulating autophagy, and activating the PI3K/Akt/mTOR signaling pathway in neurons, which may make it a promising candidate for the simultaneous treatment of VD and diabetes.
Collapse
|
11
|
Yu W, Ren C, Ji X. A review of remote ischemic conditioning as a potential strategy for neural repair poststroke. CNS Neurosci Ther 2022; 29:516-524. [PMID: 36550592 PMCID: PMC9873528 DOI: 10.1111/cns.14064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/17/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Ischemic stroke is one of the major disabling health-care problem and multiple different approaches are needed to enhance rehabilitation, in which neural repair is the structural basement. Remote ischemic conditioning (RIC) is a strategy to trigger endogenous protect. RIC has been reported to play neuroprotective role in acute stage of stroke, but the effect of RIC on repair process remaining unclear. Several studies have discovered some overlapped mechanisms RIC and neural repair performs. This review provides a hypothesis that RIC is a potential therapeutic strategy on stroke rehabilitation by evaluating the existing evidence and puts forward some remaining questions to clarify and future researches to be performed in the field.
Collapse
Affiliation(s)
- Wantong Yu
- Department of Neurology and Beijing Key Laboratory of Hypoxia Translational MedicineXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Changhong Ren
- Department of Neurology and Beijing Key Laboratory of Hypoxia Translational MedicineXuanwu Hospital, Capital Medical UniversityBeijingChina,Center of Stroke, Beijing Institute for Brain DisorderCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Department of Neurology and Beijing Key Laboratory of Hypoxia Translational MedicineXuanwu Hospital, Capital Medical UniversityBeijingChina,Center of Stroke, Beijing Institute for Brain DisorderCapital Medical UniversityBeijingChina
| |
Collapse
|
12
|
Belmonte KCD, Holmgren EB, Wills TA, Gidday JM. Epigenetic conditioning induces intergenerational resilience to dementia in a mouse model of vascular cognitive impairment. Alzheimers Dement 2022; 18:1711-1720. [PMID: 35170835 PMCID: PMC9790554 DOI: 10.1002/alz.12616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Epigenetic stimuli induce beneficial or detrimental changes in gene expression, and consequently, phenotype. Some of these phenotypes can manifest across the lifespan-and even in subsequent generations. Here, we used a mouse model of vascular cognitive impairment and dementia (VCID) to determine whether epigenetically induced resilience to specific dementia-related phenotypes is heritable by first-generation progeny. METHODS Our systemic epigenetic therapy consisted of 2 months of repetitive hypoxic "conditioning" (RHC) prior to chronic cerebral hypoperfusion in adult C57BL/6J mice. Resultant changes in object recognition memory and hippocampal long-term potentiation (LTP) were assessed 3 and 4 months later, respectively. RESULTS Hypoperfusion-induced memory/plasticity deficits were abrogated by RHC. Moreover, similarly robust dementia resilience was documented in untreated cerebral hypoperfused animals derived from RHC-treated parents. CONCLUSIONS Our results in experimental VCID underscore the efficacy of epigenetics-based treatments to prevent memory loss, and demonstrate for the first time the heritability of an induced resilience to dementia.
Collapse
Affiliation(s)
- Krystal Courtney D. Belmonte
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Eleanor B. Holmgren
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Tiffany A. Wills
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Jeff M. Gidday
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of Biochemistry and Molecular BiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| |
Collapse
|
13
|
Sun M, Jiang C, Hao X, Pang J, Chen C, Xiang W, Zhang J, Zhao S, Wang P, Geng S, Wang H, Li Y, Wang B. Long-term L-3-n-butylphthalide pretreatment attenuates ischemic brain injury in mice with permanent distal middle cerebral artery occlusion through the Nrf2 pathway. Heliyon 2022; 8:e09909. [PMID: 35874077 PMCID: PMC9305368 DOI: 10.1016/j.heliyon.2022.e09909] [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: 11/15/2021] [Revised: 02/24/2022] [Accepted: 07/04/2022] [Indexed: 10/25/2022] Open
Abstract
L-3-n-butylphthalide (NBP), which is used for treatment of mild and moderate acute ischemic stroke, exerts its effects by modulating the Nrf2 pathway. However, it has not been established whether NBP exerts its preventive effects in high-risk ischemic stroke patients through the Nrf2 pathway. We investigated whether NBP exerts its preventive effects through the Nrf2 pathway in long-term NBP pretreated dMCAO mice models. Nrf2+/+ wild-type and Nrf2-/- knockout mice were randomized into the vehicle group (equal volume vegetable oil), NBP-low-dose group (20 mg/kg) and NBP-high-dose group (60 mg/kg). The drug was administered once daily by gavage for a month. Then, a permanent distal middle cerebral artery occlusion model (dMCAO) was established after pretreatment with NBP. Neurological deficits, cerebral infarct volumes, brain water contents, activities of SOD, GSH-Px and MDA levels were determined. Further, axonal injury and demyelination, expression levels of Nrf2, HO-1 and NQO1 in ischemic brains were determined. Long-term NBP pretreatment significantly improved neurological functions, reduced cerebral infarction volumes, reduced brain water contents, increased SOD, GSH-Px activities, decreased MDA contents, reduced neurological injuries, axonal damage as well as demyelination, while increasing Nrf2, HO-1 and NQO1 mRNA as well as protein expressions in dMCAO mice models.
Collapse
Affiliation(s)
- Mingying Sun
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Neurology Academician Workstation of Baotou Central Hospital, Inner Mongolia, China
| | - Changchun Jiang
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China
| | - Xiwa Hao
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China.,Neurology Academician Workstation of Baotou Central Hospital, Inner Mongolia, China
| | - Jiangxia Pang
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China.,Neurology Academician Workstation of Baotou Central Hospital, Inner Mongolia, China
| | - Chao Chen
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China.,Neurology Academician Workstation of Baotou Central Hospital, Inner Mongolia, China
| | - Wenping Xiang
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China
| | - Jun Zhang
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China
| | - Shijun Zhao
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China
| | - Po Wang
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China
| | - Shangyong Geng
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China
| | - Hanzhang Wang
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China
| | - Yuechun Li
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China
| | - Baojun Wang
- Department of Neurology, Baotou Central Hospital, Inner Mongolia, China.,Neurological Diseases Clinical Medicine Research Center, Inner Mongolia Autonomous Region, China.,Cerebrovascular Disease Institute, Inner Mongolia Autonomous Region, China
| |
Collapse
|
14
|
AKINCI UYSAL Ç, TEMİZ REŞİTOĞLU M, GÜDEN DS, ŞENOL SP, VEZİR Ö, SUCU N, TUNÇTAN B, MALİK KU, FIRAT S. Inhibition of mTOR protects against skeletal muscle and kidney injury following hindlimb ischemia-reperfusion in rats by regulating MERK1/ERK1/2 activity. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1021518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
15
|
Baltan S, Sandau US, Brunet S, Bastian C, Tripathi A, Nguyen H, Liu H, Saugstad JA, Zarnegarnia Y, Dutta R. Identification of miRNAs That Mediate Protective Functions of Anti-Cancer Drugs During White Matter Ischemic Injury. ASN Neuro 2021; 13:17590914211042220. [PMID: 34619990 PMCID: PMC8642107 DOI: 10.1177/17590914211042220] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We have previously shown that two anti-cancer drugs, CX-4945 and MS-275, protect and preserve white matter (WM) architecture and improve functional recovery in a model of WM ischemic injury. While both compounds promote recovery, CX-4945 is a selective Casein kinase 2 (CK2) inhibitor and MS-275 is a selective Class I histone deacetylase (HDAC) inhibitor. Alterations in microRNAs (miRNAs) mediate some of the protective actions of these drugs. In this study, we aimed to (1) identify miRNAs expressed in mouse optic nerves (MONs); (2) determine which miRNAs are regulated by oxygen glucose deprivation (OGD); and (3) determine the effects of CX-4945 and MS-275 treatment on miRNA expression. RNA isolated from MONs from control and OGD-treated animals with and without CX-4945 or MS-275 treatment were quantified using NanoString nCounter® miRNA expression profiling. Comparative analysis of experimental groups revealed that 12 miRNAs were expressed at high levels in MONs. OGD upregulated five miRNAs (miR-1959, miR-501-3p, miR-146b, miR-201, and miR-335-3p) and downregulated two miRNAs (miR-1937a and miR-1937b) compared to controls. OGD with CX-4945 upregulated miR-1937a and miR-1937b, and downregulated miR-501-3p, miR-200a, miR-1959, and miR-654-3p compared to OGD alone. OGD with MS-275 upregulated miR-2134, miR-2141, miR-2133, miR-34b-5p, miR-153, miR-487b, miR-376b, and downregulated miR-717, miR-190, miR-27a, miR-1959, miR-200a, miR-501-3p, and miR-200c compared to OGD alone. Interestingly, miR-501-3p and miR-1959 were the only miRNAs upregulated by OGD, and downregulated by OGD plus CX-4945 and MS-275. Therefore, we suggest that protective functions of CX-4945 or MS-275 against WM injury maybe mediated, in part, through miRNA expression.
Collapse
Affiliation(s)
- Selva Baltan
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Selva Baltan, Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Mackenzie Hall 2140A, L459, 3181 S.W. Sam Jackson Park Rd., Portland, OR 97239, USA.
| | - Ursula S. Sandau
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Sylvain Brunet
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Chinthasagar Bastian
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Ajai Tripathi
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Hung Nguyen
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Helen Liu
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Julie A. Saugstad
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Yalda Zarnegarnia
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ranjan Dutta
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| |
Collapse
|
16
|
Xu R, He Q, Wang Y, Yang Y, Guo ZN. Therapeutic Potential of Remote Ischemic Conditioning in Vascular Cognitive Impairment. Front Cell Neurosci 2021; 15:706759. [PMID: 34413726 PMCID: PMC8370253 DOI: 10.3389/fncel.2021.706759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/29/2021] [Indexed: 12/21/2022] Open
Abstract
Vascular cognitive impairment (VCI) is a heterogeneous disease caused by a variety of cerebrovascular diseases. Patients with VCI often present with slower cognitive processing speed and poor executive function, which affects their independence in daily life, thus increasing social burden. Remote ischemic conditioning (RIC) is a non-invasive and efficient intervention that triggers endogenous protective mechanisms to generate neuroprotection. Over the past decades, evidence from basic and clinical research has shown that RIC is promising for the treatment of VCI. To further our understanding of RIC and improve the management of VCI, we summarize the evidence on the therapeutic potential of RIC in relation to the risk factors and pathobiologies of VCI, including reducing the risk of recurrent stroke, decreasing high blood pressure, improving cerebral blood flow, restoring white matter integrity, protecting the neurovascular unit, attenuating oxidative stress, and inhibiting the inflammatory response.
Collapse
Affiliation(s)
- Rui Xu
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Qianyan He
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yan Wang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
17
|
Ren C, Liu Y, Stone C, Li N, Li S, Li H, Cheng Z, Hu J, Li W, Jin K, Ji X, Ding Y. Limb Remote Ischemic Conditioning Ameliorates Cognitive Impairment in Rats with Chronic Cerebral Hypoperfusion by Regulating Glucose Transport. Aging Dis 2021; 12:1197-1210. [PMID: 34341702 PMCID: PMC8279524 DOI: 10.14336/ad.2020.1125] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/14/2020] [Indexed: 11/01/2022] Open
Abstract
Cognitive impairment is closely associated with the slowing of glucose metabolism in the brain. Glucose transport, a rate-limiting step of glucose metabolism, plays a key role in this phenomenon. Previous studies have reported that limb remote ischemic conditioning (LRIC) improves cognitive performance in rats with chronic cerebral hypoperfusion (CCH). Here, we determined whether LRIC could ameliorate cognitive impairment in rats with CCH by regulating glucose transport. A total of 170 male Sprague-Dawley rats were used. Animals subjected to permanent double carotid artery occlusion (2VO) were assigned to the control or LRIC treatment group. LRIC was applied beginning 3 days after the 2VO surgery. We found that LRIC can improve learning and memory; decrease the ratio of ADP/ATP; increase glucose content; upregulate the expression of pAMPKα, GLUT1 and GLUT3; and increase the number of GLUT1 and GLUT3 transporters in cerebral cortical neurons. The expression of GLUT1 and GLUT3 in the cortex displayed a strong correlation with learning and memory. Pearson correlation analysis showed that the levels of GLUT1 and GLUT3 are correlated with neurological function scores. All of these beneficial effects of LRIC were ablated by application of the AMPK inhibitor, dorsomorphin. In summary, LRIC ameliorated cognitive impairment in rats with CCH by regulating glucose transport via the AMPK/GLUT signaling pathway. We conclude that AMPK-mediated glucose transport plays a key role in LRIC. These data also suggest that supplemental activation of glucose transport after CCH may provide a clinically applicable intervention for improving cognitive impairment.
Collapse
Affiliation(s)
- Changhong Ren
- 1Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,5Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yuanyuan Liu
- 1Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,2Department of Endocrinology, The Affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, China
| | - Christopher Stone
- 4Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ning Li
- 1Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,5Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Sijie Li
- 1Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haiyan Li
- 1Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,5Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Zichao Cheng
- 1Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,3Department of Rehabilitation Medicine, Affiliated 3201 Hospital of Xi'an Jiaotong University School of Medicine, Hanzhong, China
| | - Jiangnan Hu
- 6Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Weiguang Li
- 1Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kunlin Jin
- 7Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Xunming Ji
- 5Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- 1Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,4Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
18
|
Shikonin Attenuates Chronic Cerebral Hypoperfusion-Induced Cognitive Impairment by Inhibiting Apoptosis via PTEN/Akt/CREB/BDNF Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5564246. [PMID: 34211568 PMCID: PMC8205575 DOI: 10.1155/2021/5564246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/13/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022]
Abstract
Shikonin (SK) exerts neuroprotective effects; however, to date, its protective effect against chronic cerebral hypoperfusion- (CCH-) induced vascular dementia (VaD) has not been investigated. Therefore, the current study investigated whether SK could mitigate the cognitive deficits caused by CCH. The effects of SK treatment on the PTEN/Akt/CREB/BDNF signaling pathway and apoptosis in hippocampal neurons were examined in a rat model of VaD established via bilateral common carotid artery occlusion (BCCAO). Fifty-two rats were randomly divided into 4 groups: sham, vehicle, SK-L (10 mg/kg SK per day), and SK-H (25 mg/kg SK per day). SK was regularly administered by gavage for 2 weeks. The results of the water maze test revealed that the escape latency in the vehicle group was significantly longer than that in the sham group, and rats in the vehicle group spent a smaller proportion of time in the target quadrant than those in the sham group. SK treatment reduced the escape latencies and increased the proportion of time spent in the target quadrant. Nissl staining showed morphological damage in the CA1 areas of the hippocampus in the vehicle group. SK treatment alleviated the injuries to hippocampal neurons. Western blot analysis showed higher p-PTEN and lower p-Akt, p-CREB, and BDNF expression in the vehicle group than in the sham group. SK administration reversed the upregulation of p-PTEN and the downregulation of p-Akt, p-CREB, and BDNF. The number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling- (TUNEL-) positive cells in the hippocampal CA1 region of the vehicle group was significantly increased. Treatment with SK decreased the number of positive cells. Furthermore, as marker proteins of apoptosis, bcl-2 expression was decreased and bax expression was increased; thus, the ratio of bcl-2/bax was decreased in the vehicle group. SK treatment upregulated the expression of bcl-2 and downregulated the expression of bax, thereby elevating the bcl-2/bax ratio. Moreover, the aforementioned effects of SK were dose-dependent. The effect of 25 mg/kg per day was more obvious than that of 10 mg/kg per day. In conclusion, SK inhibited hippocampal neuronal apoptosis to protect against CCH-induced injury by regulating the PTEN/Akt/CREB/BDNF signaling pathway, consequently improving cognitive impairment.
Collapse
|
19
|
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: 17] [Impact Index Per Article: 4.3] [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
|
20
|
Li C, Wang Y, Yan XL, Guo ZN, Yang Y. Pathological changes in neurovascular units: Lessons from cases of vascular dementia. CNS Neurosci Ther 2021; 27:17-25. [PMID: 33423390 PMCID: PMC7804924 DOI: 10.1111/cns.13572] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular dementia (VD) is the second leading cause of dementia after Alzheimer's disease (AD). The decrease of cerebral blood flow (CBF) to different degrees is one of the main causes of VD. Neurovascular unit (NVU) is a vessel‐centered concept, emphasizing all the cellular components play an integrated role in maintaining the normal physiological functions of the brain. More and more evidence shows that reduced CBF causes a series of changes in NVU, such as impaired neuronal function, abnormal activation of glial cells, and changes in vascular permeability, all of which collectively play a role in the pathogenesis of VD. In this paper, we review NVU changes as CBF decreases, focusing on each cellular component of NVU. We also highlight remote ischemic preconditioning as a promising approach for VD prevention and treatment from the NVU perspective of view.
Collapse
Affiliation(s)
- Chao Li
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yan Wang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Xiu-Li Yan
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
21
|
Li M, Meng N, Guo X, Niu X, Zhao Z, Wang W, Xie X, Lv P. Dl-3-n-Butylphthalide Promotes Remyelination and Suppresses Inflammation by Regulating AMPK/SIRT1 and STAT3/NF-κB Signaling in Chronic Cerebral Hypoperfusion. Front Aging Neurosci 2020; 12:137. [PMID: 32581761 PMCID: PMC7296049 DOI: 10.3389/fnagi.2020.00137] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Demyelination in vascular dementia (VD) is partly attributable to inflammation induced by chronic cerebral hypoperfusion (CCH). Remyelination contributes to the recovery of cognitive impairment by inducing the proliferation and differentiation of oligodendrocyte progenitor cells. It was previously reported that Dl-3-n-butylphthalide (NBP) promotes cognitive improvement. However, whether NBP can stimulate remyelination and suppress inflammation after CCH remains unclear. To answer this question, the present study investigated the effects of NBP on remyelination in a rat model of CCH established by bilateral carotid artery occlusion. Functional recovery was evaluated with the Morris water maze (MWM) test, and myelin integrity, regeneration of mature oligodendrocytes, and inhibition of astrocyte proliferation were assessed by immunohistochemistry and histologic analysis. Additionally, activation of 5′ AMP-activated protein kinase (AMPK)/Sirtuin (SIRT)1 and Signal transducer and activator of transcription (STAT)3/nuclear factor (NF)-κB signaling pathways was evaluated by western blotting. The results showed that NBP treatment improved memory and learning performance in CCH rats, which was accompanied by increased myelin integrity and oligodendrocyte regeneration, and reduced astrocyte proliferation and inflammation. Additionally, NBP induced the activation of AMPK/SIRT1 signaling while inhibiting the STAT3/NF-κB pathway. These results indicate that NBP alleviates cognitive impairment following CCH by promoting remyelination and suppressing inflammation via modulation of AMPK/SIRT1 and STAT3/NF-κB signaling.
Collapse
Affiliation(s)
- Meixi Li
- Department of Neurology, Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Nan Meng
- Department of Neurology, Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Xin Guo
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Xiaoli Niu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Zhongmin Zhao
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Wei Wang
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Xiaohua Xie
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
22
|
Wang DP, Chen SH, Wang D, Kang K, Wu YF, Su SH, Zhang YY, Hai J. Neuroprotective effects of andrographolide on chronic cerebral hypoperfusion-induced hippocampal neuronal damage in rats possibly via PTEN/AKT signaling pathway. Acta Histochem 2020; 122:151514. [PMID: 32019701 DOI: 10.1016/j.acthis.2020.151514] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/02/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023]
Abstract
To explore the potential effects of andrographolide on chronic cerebral hypoperfusion (CCH)-induced neuronal damage as well as the underlying mechanisms. Rat CCH model was established by 2-vessel occlusion (2VO). The CCH rats received andrographolide treatment for 4 weeks. The neuron loss was detected by using neuronal nuclei (NeuN) immunofluorescent staining. The expression levels of phospho-phosphatase and tensin homolog deleted on chromosome ten (p-PTEN), protein kinase B (AKT), p-AKT, and cysteinyl aspartate specific proteinase-3 (Caspase-3) proteins were accessed by Western blotting. Moreover, the neuronal apoptosis of hippocampus tissues was detected via terminal deoxynucleotidyl transferase- mediated dUTP nick end labeling (TUNEL) staining. CCH reduced the number of NeuN-positive cells, while the number was significant increased after andrographolide treatment. CCH increased the proteins expression level of p-PTEN, Caspase-3, and decreased the p-AKT, which were reversed by andrographolide treatment. Furthermore, andrographolide treatment also down-regulated CCH-induced TUNEL-apoptosis rate. Our results suggest that the PTEN/AKT pathway may be modulated by andrographolide and the damaging effects of CCH on hippocampus may be ameliorated by andrographolide treatment. Andrographolide may act as a potential therapeutic approach for chronic ischemic insults.
Collapse
|
23
|
Zhang LY, Pan J, Mamtilahun M, Zhu Y, Wang L, Venkatesh A, Shi R, Tu X, Jin K, Wang Y, Zhang Z, Yang GY. Microglia exacerbate white matter injury via complement C3/C3aR pathway after hypoperfusion. Theranostics 2020; 10:74-90. [PMID: 31903107 PMCID: PMC6929610 DOI: 10.7150/thno.35841] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/08/2019] [Indexed: 12/22/2022] Open
Abstract
Microglial activation participates in white matter injury after cerebral hypoperfusion. However, the underlying mechanism is unclear. Here, we explore whether activated microglia aggravate white matter injury via complement C3-C3aR pathway after chronic cerebral hypoperfusion. Methods: Adult male Sprague-Dawley rats (n = 80) underwent bilateral common carotid artery occlusion for 7, 14, and 28 days. Cerebral vessel density and blood flow were examined by synchrotron radiation angiography and three-dimensional arterial spin labeling. Neurobehavioral assessments, CLARITY imaging, and immunohistochemistry were performed to evaluate activation of microglia and C3-C3aR pathway. Furthermore, C3aR knockout mice were used to establish the causal relationship of C3-C3aR signaling on microglia activation and white matter injury after hypoperfusion. Results: Cerebral vessel density and blood flow were reduced after hypoperfusion (p<0.05). Spatial learning and memory deficits and white matter injury were shown (p<0.05). These impairments were correlated with aberrant microglia activation and an increase in the number of reactive microglia adhering to and phagocytosed myelin in the hypoperfusion group (p<0.05), which were accompanied by the up-regulation of complement C3 and its receptors C3aR (p<0.05). Genetic deletion of C3ar1 significantly inhibited aberrant microglial activation and reversed white matter injury after hypoperfusion (p<0.05). Furthermore, the C3aR antagonist SB290157 decreased the number of microglia adhering to myelin (p<0.05), attenuated white matter injury and cognitive deficits in chronic hypoperfusion rats (p<0.05). Conclusions: Our results demonstrated that aberrant activated microglia aggravate white matter injury via C3-C3aR pathway during chronic hypoperfusion. These findings indicate C3aR plays a critical role in mediating neuroinflammation and white matter injury through aberrant microglia activation, which provides a novel therapeutic target for the small vessel disease and vascular dementia.
Collapse
|
24
|
Rossino MG, Dal Monte M, Casini G. Relationships Between Neurodegeneration and Vascular Damage in Diabetic Retinopathy. Front Neurosci 2019; 13:1172. [PMID: 31787868 PMCID: PMC6856056 DOI: 10.3389/fnins.2019.01172] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022] Open
Abstract
Diabetic retinopathy (DR) is a common complication of diabetes and constitutes a major cause of vision impairment and blindness in the world. DR has long been described exclusively as a microvascular disease of the eye. However, in recent years, a growing interest has been focused on the contribution of neuroretinal degeneration to the pathogenesis of the disease, and there are observations suggesting that neuronal death in the early phases of DR may favor the development of microvascular abnormalities, followed by the full manifestation of the disease. However, the mediators that are involved in the crosslink between neurodegeneration and vascular changes have not yet been identified. According to our hypothesis, vascular endothelial growth factor (VEGF) could probably be the most important connecting link between the death of retinal neurons and the occurrence of microvascular lesions. Indeed, VEGF is known to play important neuroprotective actions; therefore, in the early phases of DR, it may be released in response to neuronal suffering, and it would act as a double-edged weapon inducing both neuroprotective and vasoactive effects. If this hypothesis is correct, then any retinal stress causing neuronal damage should be accompanied by VEGF upregulation and by vascular changes. Similarly, any compound with neuroprotective properties should also induce VEGF downregulation and amelioration of the vascular lesions. In this review, we searched for a correlation between neurodegeneration and vasculopathy in animal models of retinal diseases, examining the effects of different neuroprotective substances, ranging from nutraceuticals to antioxidants to neuropeptides and others and showing that reducing neuronal suffering also prevents overexpression of VEGF and vascular complications. Taken together, the reviewed evidence highlights the crucial role played by mediators such as VEGF in the relationship between retinal neuronal damage and vascular alterations and suggests that the use of neuroprotective substances could be an efficient strategy to prevent the onset or to retard the development of DR.
Collapse
Affiliation(s)
| | - Massimo Dal Monte
- Department of Biology, University of Pisa, Pisa, Italy.,Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| | - Giovanni Casini
- Department of Biology, University of Pisa, Pisa, Italy.,Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| |
Collapse
|
25
|
Wang A, Wang R, Cui D, Huang X, Yuan L, Liu H, Fu Y, Liang L, Wang W, He Q, Shi C, Guan X, Teng Z, Zhao G, Li Y, Gao Y, Han H. The Drainage of Interstitial Fluid in the Deep Brain is Controlled by the Integrity of Myelination. Aging Dis 2019; 10:937-948. [PMID: 31595193 PMCID: PMC6764732 DOI: 10.14336/ad.2018.1206] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/26/2018] [Indexed: 12/17/2022] Open
Abstract
In searching for the drainage route of the interstitial fluid (ISF) in the deep brain, we discovered a regionalized ISF drainage system as well as a new function of myelin in regulating the drainage. The traced ISF from the caudate nucleus drained to the ipsilateral cortex along myelin fiber tracts, while in the opposite direction, its movement to the adjacent thalamus was completely impeded by a barrier structure, which was identified as the converged, compact myelin fascicle. The regulating and the barrier effects of myelin were unchanged in AQP4-knockout rats but were impaired as the integrity of boundary structure of drainage system was destroyed in a demyelinated rat model. We thus proposed that the brain homeostasis was maintained within each ISF drainage division locally, rather than across the brain as a whole. A new brain division system and a new pathogenic mechanism of demyelination are therefore proposed.
Collapse
Affiliation(s)
- Aibo Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Rui Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Dehua Cui
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Xinrui Huang
- Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Lan Yuan
- Peking University Medical and Health Analysis Center, Peking University Health Science Center, Beijing, China.
| | - Huipo Liu
- Institute of Applied Physics and Computational Mathematics, Beijing, China.
| | - Yu Fu
- Department of Neurology, Peking University Third Hospital, Beijing, China.
| | - Lei Liang
- Department of Medical Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Wei Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Qingyuan He
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Chunyan Shi
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Xiangping Guan
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Ze Teng
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Guomei Zhao
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Yuanyuan Li
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Yajuan Gao
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| |
Collapse
|
26
|
Hu L, Zhang H, Wang B, Ao Q, Shi J, He Z. MicroRNA-23b alleviates neuroinflammation and brain injury in intracerebral hemorrhage by targeting inositol polyphosphate multikinase. Int Immunopharmacol 2019; 76:105887. [PMID: 31536904 DOI: 10.1016/j.intimp.2019.105887] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/21/2019] [Accepted: 09/05/2019] [Indexed: 12/27/2022]
Abstract
Neuroinflammation plays a critical role in the pathogenesis of intracerebral hemorrhage (ICH), contributing to detrimental brain injury and neurological function deficits. MicroRNA-23b (miR-23b) exerts anti-inflammatory effects in many diseases and is downregulated in patients with ICH. This study aimed to evaluate the involvement of miR-23b in ICH models in vivo and in vitro, using basal ganglia injection of collagenase type VII in rats and hemin stimulation for cells, respectively. Exogenous overexpression of miR-23b by transfection with lentivirus-miR-23b (LV-miR-23b) or miR-23b mimics was evaluated by RT-qPCR. In this study, we found miR-23b was downregulated in the ICH models and its overexpression effectively alleviated neurological deficits, brain edema, hematoma area, and neuronal apoptosis in ICH rats. Western blotting for neuroinflammation markers and immunofluorescence staining for microglial activation demonstrated that miR-23b could alleviate neuroinflammation in ICH in vivo. We also performed an in vitro mechanism study using BV2 microglial cells and HT22 neuronal cell lines to explore how miR-23b modulates neuroinflammation and neuronal protection after ICH. We found that miR-23b significantly decreased hemin-stimulated inflammation response in BV2 cells and attenuated co-cultured HT22 neuronal cell death. Additionally, we verified that miR-23b suppressed inflammation in BV2 cells by targeting inositol polyphosphate multikinase (IPMK) and that autophagy regulation through the Akt/mTOR pathway was involved in miR-23b-regulated inflammation after ICH. Our study illustrated that miR-23b played a protective role in ICH through inhibiting neuroinflammation by targeting IPMK; this mechanism may be related to the regulation of the Akt/mTOR autophagy pathway, making it a potential target for ICH treatment.
Collapse
Affiliation(s)
- Liuting Hu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110000,People's Republic of China
| | - Heyu Zhang
- Dapartment of Neurology, The First Affiliated Hospital Sun Yat-sen University,Guangzhou 510080,People's Republic of China
| | - Bingyang Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110000,People's Republic of China
| | - Qiang Ao
- Department of Tissue Engineering, China Medical University, Shenyang 110122, People's Republic of China
| | - Jing Shi
- Department of Neurology, Dandong Central Hospital, Dandong 118002,People's Republic of China
| | - Zhiyi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110000,People's Republic of China.
| |
Collapse
|
27
|
Chen N, Caruso C, Alonso A, Derebail VK, Kshirsagar AV, Sharrett AR, Key NS, Gottesman RF, Grove ML, Bressler J, Boerwinkle E, Windham BG, Mosley TH, Hyacinth HI. Association of sickle cell trait with measures of cognitive function and dementia in African Americans. eNeurologicalSci 2019; 16:100201. [PMID: 31384675 PMCID: PMC6661502 DOI: 10.1016/j.ensci.2019.100201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/21/2019] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE The incidence and prevalence of cognitive decline and dementia are significantly higher among African Americans compared with non-Hispanic Whites. The aim of this study was to determine whether inheritance of the sickle cell trait (SCT) i.e. heterozygosity for the sickle cell mutation increases the risk of cognitive decline or dementia Among African Americans. METHODS We studied African American participants enrolled in the Atherosclerosis Risk in Communities study. SCT genotype at baseline and outcome data from cognitive assessments at visits 2, 4 and 5, and an MRI performed at visit 5 were analyzed for the association between SCT and risk of cognitive impairment and/or dementia. RESULTS There was no significant difference in risk factors profile between participants with SCT (N = 176) and those without SCT (N = 2532). SCT was not independently associated with a higher prevalence of global or domain-specific cognitive impairment at baseline or with more rapid cognitive decline. Participants with SCT had slightly lower incidence of dementia (HR = 0.63 [0.38, 1.05]). On the other hand, SCT seems to interact with the apolipoprotein E ε4 risk allele resulting in poor performance on digit symbol substitution test at baseline (z-score = -0.08, Pinteraction = 0.05) and over time (z-score = -0.12, Pinteraction = 0.04); and with diabetes mellitus leading to a moderately increased risk of dementia (HR = 2.06 [0.89, 4.78], Pinteraction = 0.01). CONCLUSIONS SCT was not an independent risk factor for prevalence or incidence of cognitive decline or dementia, although it may interact with and modify other putative risk factors for cognitive decline and dementia.
Collapse
Affiliation(s)
- Nemin Chen
- Department of Epidemiology, University of Pittsburg, Pittsburg, PA, United States of America
| | - Christina Caruso
- Aflac Cancer and Blood Disorder Center of Children's Healthcare of Atlanta, Emory Department of Pediatrics, Atlanta, GA, United States of America
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - Vimal K. Derebail
- UNC Kidney Center, Division of Nephrology and Hypertension, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Abhijit V. Kshirsagar
- UNC Kidney Center, Division of Nephrology and Hypertension, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - A. Richey Sharrett
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Nigel S. Key
- University of North Carolina, Department of Medicine, Chapel Hill, NC, United States of America
| | - Rebecca F. Gottesman
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States of America
| | - Megan L. Grove
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, United States of America
- Human Genome Sequencing Center at Baylor College of Medicine, Houston, TX, United States of America
| | - B. Gwen Windham
- University of Mississippi Medical Center, Department of Medicine/Geriatrics, Jackson, MS, United States of America
| | - Thomas H. Mosley
- MIND Center, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Hyacinth I. Hyacinth
- Aflac Cancer and Blood Disorder Center of Children's Healthcare of Atlanta, Emory Department of Pediatrics, Atlanta, GA, United States of America
| |
Collapse
|
28
|
Role of exosomes induced by remote ischemic preconditioning in neuroprotection against cerebral ischemia. Neuroreport 2019; 30:834-841. [DOI: 10.1097/wnr.0000000000001280] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Chen C, Zhang H, Xu H, Xue R, Zheng Y, Wu T, Lian Y. Harpagoside Rescues the Memory Impairments in Chronic Cerebral Hypoperfusion Rats by Inhibiting PTEN Activity. J Alzheimers Dis 2019; 63:445-455. [PMID: 29614669 DOI: 10.3233/jad-171170] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Vascular dementia (VaD) is the second most common dementia worldwide. Unlike Alzheimer's disease, VaD does not yet have effective therapeutic drugs. Harpagoside is the most important component extracted from Harpagophytum procumbens, a traditional Chinese medicine that has been widely used. The neuroprotective effects of harpagoside have been studied in Aβ- and MPTP-induced neurotoxicity. However, whether harpagoside is protective against VaD is not clear. In this study, with the use of chronic cerebral hypoperfusion rats, a well-known VaD model, we demonstrated that chronic administration (two months) of harpagoside was able to restore both the spatial learning/memory and fear memory impairments. Importantly, the protective effects of harpagoside were not due to alterations in the physiological conditions, metabolic parameters, or locomotor abilities of the rats. Meanwhile, we found that harpagoside suppressed the overactivation of PTEN induced by CCH by enhancing PTEN phosphorylation. Furthermore, harpagoside elevated the activity of Akt and inhibited the activity of GSK-3β, downstream effectors of PTEN. Overall, our study suggested that harpagoside treatment might be a potential therapeutic drug targeting the cognitive impairments of VaD.
Collapse
Affiliation(s)
- Chen Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Haifeng Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Hongliang Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Yake Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Tianwen Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, People's Republic of China
| |
Collapse
|
30
|
You J, Feng L, Bao L, Xin M, Ma D, Feng J. Potential Applications of Remote Limb Ischemic Conditioning for Chronic Cerebral Circulation Insufficiency. Front Neurol 2019; 10:467. [PMID: 31130914 PMCID: PMC6509171 DOI: 10.3389/fneur.2019.00467] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic cerebral circulation insufficiency (CCCI) refers to a chronic decrease in cerebral blood perfusion, which may lead to cognitive impairment, psychiatric disorders such as depression, and acute ischemic stroke. Remote limb ischemic conditioning (RLIC), in which the limbs are subjected to a series of transient ischemic attacks, can activate multiple endogenous protective mechanisms to attenuate fatal ischemic injury to distant organs due to acute ischemia, such as ischemic stroke. Recent studies have also reported that RLIC can alleviate dysfunction in distant organs caused by chronic, non-fatal reductions in blood supply (e.g., CCCI). Indeed, research has indicated that RLIC may exert neuroprotective effects against CCCI through a variety of potential mechanisms, including attenuated glutamate excitotoxicity, improved endothelial function, increased cerebral blood flow, regulation of autophagy and immune responses, suppression of apoptosis, the production of protective humoral factors, and attenuated accumulation of amyloid-β. Verification of these findings is necessary to improve prognosis and reduce the incidence of acute ischemic stroke/cognitive impairment in patients with CCCI.
Collapse
Affiliation(s)
- Jiulin You
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Liangshu Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Liyang Bao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Meiying Xin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
31
|
Ding J, Zhou D, Liu C, Pan L, Ya J, Ding Y, Ji X, Meng R. Normobaric oxygen: a novel approach for treating chronic cerebral circulation insufficiency. Clin Interv Aging 2019; 14:565-570. [PMID: 30936686 PMCID: PMC6421875 DOI: 10.2147/cia.s190984] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Chronic cerebral circulation insufficiency (CCCI) is viewed as an alarming state induced by long-term reduction in cerebral perfusion, which is associated with neurological deficits and high risk of stroke occurrence or recurrence. CCCI accounts for a large proportion of both outpatients and inpatients with cerebrovascular diseases, while management of CCCI remains a formidable challenge to clinicians. Normobaric oxygen (NBO) is an adjuvant hyperoxygenation intervention supplied with one atmosphere pressure (1 ATA =101.325 kPa). A plethora of studies have demonstrated the efficacy of NBO on the penumbra in acute stroke. NBO has been shown to increase the oxygen pressure, raise the intracranial blood flow, protect blood–brain barrier and enhance neuroprotective effects. As similar underlying mechanisms are shared by the penumbra in stroke and the ischemic–hypoxic brain tissues in CCCI, we speculate that NBO may serve as a promising therapeutic strategy for attenuating short-term symptoms or improving long-term clinical outcomes among patients with CCCI. Due to the scant research exploring the efficacy and safety of NBO for treating CCCI so far, both experimental and clinical studies are warranted to verify our hypothesis in the future.
Collapse
Affiliation(s)
- Jiayue Ding
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China, .,Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China, .,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China,
| | - Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China, .,Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China, .,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China,
| | - Cheng Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China, .,Department of Neurology, Yongxin People's Hospital, Ji'an 343400, China
| | - Liqun Pan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China, .,Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China, .,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China,
| | - Jingyuan Ya
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China, .,Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China, .,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China,
| | - Yuchuan Ding
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China, .,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xunming Ji
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China, .,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China, .,Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China, .,Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China, .,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China,
| |
Collapse
|
32
|
Zhou D, Ding J, Ya J, Pan L, Bai C, Guan J, Wang Z, Jin K, Yang Q, Ji X, Meng R. Efficacy of remote ischemic conditioning on improving WMHs and cognition in very elderly patients with intracranial atherosclerotic stenosis. Aging (Albany NY) 2019; 11:634-648. [PMID: 30689549 PMCID: PMC6366980 DOI: 10.18632/aging.101764] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 01/05/2019] [Indexed: 04/10/2023]
Abstract
Our previous study revealed that remote ischemic conditioning (RIC) reduced the incidence of stroke or TIA in octo- and nonagenarians with intracranial atherosclerotic stenosis (ICAS). Herein, we aimed to investigate whether RIC would influence the progression of white matter hyperintensities (WMHs) and cognitive impairment in the same group of patients. Fifty-eight patients with ICAS were randomly assigned in a 1:1 ratio to receive standard medical treatment with RIC (n=30) versus sham-RIC (n=28). The RIC protocol consisted of 5 cycles of alternating 5-min ischemia and 5-min reperfusion applied in the bilateral upper arms twice daily for 300 days. The efficacy outcomes included WMHs change on T2 FLAIR sequences, estimated by the Fazekas scale and Scheltens scale, cognitive change as assessed by the MMSE and MoCA, and some clinical symptoms within 300-day follow-up. Compared with the baseline, RIC treatment significantly reduced Fazekas and Scheltens scores at both 180-day (both p<0.05) and 300-day (both p<0.01) follow-ups, whereas no such reduction was observed in the control group. In the RIC group, Fazekas scores were significantly lower at 300-day follow-up (p<0.001) while Scheltens scores were significantly lower at both 180-day and 300-day follow-ups (both p<0.001), as compared with the control group. There were statistically significant between-group differences in the overall MMSE or MoCA scores, favoring RIC at 180-day and 300-day follow-ups (all p<0.05). RIC may serve as a promising adjunctive to standard medical therapy for preventing the progression of WMHs and ameliorating cognitive impairment in very elderly patients with ICAS.
Collapse
Affiliation(s)
- Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Equal contribution
| | - Jiayue Ding
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Equal contribution
| | - Jingyuan Ya
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liqun Pan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chaobo Bai
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jingwei Guan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhongao Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kexin Jin
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qi Yang
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Li F, Yang Z, Stone C, Ding JY, Previch L, Shen J, Ji Y, Geng X, Ding Y. Phenothiazines Enhance the Hypothermic Preservation of Liver Grafts: A Pilot in Vitro Study. Cell Transplant 2019; 28:318-327. [PMID: 30666889 PMCID: PMC6425111 DOI: 10.1177/0963689718824559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In vitro liver conservation is an issue of ongoing critical importance in graft transplantation. In this study, we investigated the possibility of augmenting the standard pre-transplant liver conservation protocol (University of Wisconsin (UW) cold solution) with the phenothiazines chlorpromazine and promethazine. Livers from male Sprague-Dawley rats were preserved either in UW solution alone, or in UW solution plus either 2.4, 3.6, or 4.8 mg chlorpromazine and promethazine (C+P, 1:1). The extent of liver injury following preservation was determined by alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, the ratio of AST/ALT, morphological changes as assessed by hematoxylin-eosin staining, apoptotic cell death as determined by ELISA, and by expression of the apoptotic regulatory proteins BAX and Bcl-2. Levels of glucose (GLU) and lactate dehydrogenase (LDH) in the preservation liquid were determined at 3, 12, and 24 h after incubation to assess glucose metabolism. Oxidative stress was assessed by levels of superoxide dismutase (SOD), reactive oxygen species (ROS), and malondialdehyde (MDA), and inflammatory cytokine expression was evaluated with Western blotting. C+P augmentation induced significant reductions in ALT and AST activities; the AST/ALT ratio; as well as in cellular swelling, vacuolar degeneration, apoptosis, and BAX expression. These changes were associated with lowered levels of GLU and LDH; decreased expression of SOD, MDA, ROS, TNF-α, and IL-1β; and increased expression of Bcl-2. We conclude that C+P augments hypothermic preservation of liver tissue by protecting hepatocytes from ischemia-induced oxidative stress and metabolic dysfunction. This result provides a basis for improvement of the current preservation strategy, and thus for the development of a more effective graft conservation method.
Collapse
Affiliation(s)
- Fengwu Li
- 1 China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zhiying Yang
- 2 Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Christopher Stone
- 3 Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jamie Y Ding
- 3 Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lauren Previch
- 3 Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jiamei Shen
- 1 China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,3 Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yu Ji
- 4 Department of General Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- 1 China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- 3 Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
34
|
Ren C, Wu H, Li D, Yang Y, Gao Y, Jizhang Y, Liu D, Ji X, Zhang X. Remote Ischemic Conditioning Protects Diabetic Retinopathy in Streptozotocin-induced Diabetic Rats via Anti-Inflammation and Antioxidation. Aging Dis 2018; 9:1122-1133. [PMID: 30574423 PMCID: PMC6284762 DOI: 10.14336/ad.2018.0711] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/11/2018] [Indexed: 12/13/2022] Open
Abstract
Ischemic conditioning inhibits oxidative stress and inflammatory response in diabetes. However, whether limb remote ischemic conditioning (LRIC) has beneficial effects on diabetic retinopathy (DR) remains unknown. This study aims to investigate the protective effects of LRIC in retinal ganglion cell in streptozotocin (STZ) induced Type 1 diabetic rats. A total of 48 healthy male Sprague-Dawley (200-220g) rats were randomly assigned to the normal group, normal+LRIC group, diabetes mellitus (DM) group and DM+LRIC group. Streptozotocin (STZ, 60 mg/kg) was intraperitoneally injected into the rats to establish the diabetic model. LRIC was conducted by tightening a tourniquet around the upper thigh and releasing for three cycles daily (10 mins x 3 cycles). Retinas were harvested after 12 weeks of LRIC treatment for histopathologic, Western blot and ELISA analysis. Plasma were collected at the same time for ELISA analysis. LRIC alleviated diabetic retinopathy symptoms as evidenced by the increased number of retinal ganglion cells (P<0.01) and decreased glial fibrillary acidic protein (GFAP) expression level (P<0.01) in the rat retina. LRIC in DM rats exhibited anti-inflammatory and antioxidative effects as confirmed by the down-regulation of pro-inflammatory cytokine: interleukin-6 (IL-6), and the up-regulation of antioxidants: superoxide dismutase (SOD), and glutathione (GSH)/oxidized glutathione (GSSG). Furthermore, LRIC significantly downregulated VEGF protein expression in the retina (P<0.01). These results suggest that the antioxidative and anti-inflammatory activities of LRIC may be important mechanisms involved in the protective effect of LRIC in STZ-induced diabetic rats.
Collapse
Affiliation(s)
- Changhong Ren
- 1Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.,5Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| | - Hang Wu
- 1Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.,2Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dongjie Li
- 1Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.,2Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yong Yang
- 3Department of Herbal Formula Science Medicine, Chinese Medicine College, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Gao
- 1Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.,2Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yunneng Jizhang
- 4Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Dachuan Liu
- 2Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- 1Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.,5Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| | - Xuxiang Zhang
- 1Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.,2Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
35
|
Ren C, Li N, Li S, Han R, Huang Q, Hu J, Jin K, Ji X. Limb Ischemic Conditioning Improved Cognitive Deficits via eNOS-Dependent Augmentation of Angiogenesis after Chronic Cerebral Hypoperfusion in Rats. Aging Dis 2018; 9:869-879. [PMID: 30271664 PMCID: PMC6147592 DOI: 10.14336/ad.2017.1106] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/06/2017] [Indexed: 12/27/2022] Open
Abstract
Intracranial and extracranial arterial stenosis, the primary cause of chronic cerebral hypoperfusion (CCH), is a critical reason for the pathogenesis of vascular dementia and Alzheimer’s disease characterized by cognitive impairments. Our previous study demonstrated that limb remote ischemic conditioning (LRIC) improved cerebral perfusion in intracranial arterial stenosis patients. The current study aimed to test whether LRIC promotes angiogenesis and increases phosphorylated endothelial nitric oxide synthase (p-eNOS) activity in CCH rat model. Adult male Sprague-Dawley rats were randomly assigned to three different groups: sham group, bilateral carotid artery occlusion (2VO) group and 2VO+LRIC group. Cerebral Blood Flow (CBF) was measured with laser speckle contrast imager at 4 weeks. Cognitive testing was performed at four and six weeks after 2VO surgery. We demonstrated that LRIC treatment increased cerebral perfusion and improved the CCH induced spatial learning and memory impairment. Immunohistochemistry confirmed that LRIC prevented cell death in the CA1 region, and increased the number of vessels and angiogenesis in the hippocampus after 2VO. Western blot analysis shows that LRIC therapy significantly increased p-eNOS expression in the hippocampus when compared with 2VO rats. Moreover, eNOS inhibitor reduced the effect of LRIC on angiogenesis in the hippocampus and spatial learning and memory function. Our data suggested that LRIC promoted angiogenesis, which is mediated, in part, by eNOS/NO.
Collapse
Affiliation(s)
- Changhong Ren
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, TX Texas 76107, USA.,3Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Ning Li
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,5Department of Neurobiology, Capital Medical University, Beijing 10069, China
| | - Sijie Li
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Rongrong Han
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Qingjian Huang
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, TX Texas 76107, USA.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Jiangnan Hu
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, TX Texas 76107, USA
| | - Kunlin Jin
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, TX Texas 76107, USA
| | - Xunming Ji
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,3Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| |
Collapse
|
36
|
Zhou W, Wang J, Qi Q, Feng Z, Huang B, Chen A, Zhang D, Li W, Zhang Q, Bjerkvig R, Li X, Wang J. Matrine induces senescence of human glioblastoma cells through suppression of the IGF1/PI3K/AKT/p27 signaling pathway. Cancer Med 2018; 7:4729-4743. [PMID: 30079478 PMCID: PMC6143938 DOI: 10.1002/cam4.1720] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Matrine, a traditional Chinese medicine, has recently been shown to have antitumor properties in diverse cancer cells. Here, we explored the effect of matrine on human glioblastoma multiforme (GBM) cells. METHODS Glioblastoma multiforme cell lines were treated with matrine to assess proliferation and viability using EdU and CCK8 assays. SA-β-gal assays were used to evaluate cellular senescence, and a cytokine array and ELISA assay were used to screen for secreted cytokines altered in GBM cells after matrine treatment. Immunohistochemistry and Western blot analysis were performed to evaluate protein levels in matrine-treated cell lines and in samples obtained from orthotopic xenografts. Specific activators of AKT and IGF1 were used to identify the pathways mediating the effect. RESULTS Matrine potently inhibited growth of GBM cell lines in vitro. Based on in situ assays, growth arrest induced by matrine was primarily achieved through induction of cellular senescence. Matrine treatment led to decreased expression of proteins involved in promoting cell growth, IGF1, PI3K, and pAKT. Exposure of cells to a small molecule activating AKT (SC79) and recombinant IGF1 led to a reduced number of senescent SA-β-gal-positive cells in the presence of matrine. Finally, matrine inhibited growth of orthotopic xenografts established from luciferase-stable-U251 or luciferase-stable-P3 cells and prolonged overall survival in mice. CONCLUSIONS These results indicated that matrine arrested cell growth through inhibition of IGF1/PI3K/AKT signaling. Matrine warrants further investigation as a potential therapy in the treatment of patients with GBM.
Collapse
Affiliation(s)
- Wenjing Zhou
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Jiwei Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Qichao Qi
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Zichao Feng
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Qing Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Rolf Bjerkvig
- Department of Biomedicine, K G Jebsen Brain Tumor Research Center, University of Bergen, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute, Key Laboratory of Brain Functional Remodeling, Shandong University, Jinan, Shandong, China.,Department of Biomedicine, K G Jebsen Brain Tumor Research Center, University of Bergen, Bergen, Norway
| |
Collapse
|
37
|
Cerebral ischemia induces angiogenesis in the peri-infarct regions via Notch1 signaling activation. Exp Neurol 2018; 304:30-40. [DOI: 10.1016/j.expneurol.2018.02.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/13/2018] [Accepted: 02/21/2018] [Indexed: 12/11/2022]
|
38
|
Zhao W, Li S, Ren C, Meng R, Ji X. Chronic Remote Ischemic Conditioning May Mimic Regular Exercise:Perspective from Clinical Studies. Aging Dis 2018; 9:165-171. [PMID: 29392091 PMCID: PMC5772854 DOI: 10.14336/ad.2017.1015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/15/2017] [Indexed: 11/01/2022] Open
Abstract
Chronic remote ischemic conditioning (RIC), particularly long-term repeated RIC, has been applied in clinical trials with the expectation that it could play its protective roles for protracted periods. In sports medicine, chronic RIC has also been demonstrated to improve exercise performance, akin to improvements seen with regular exercise training. Therefore, chronic RIC may mimic regular exercise, and they may have similar underlying mechanisms. In this study, we explored the common underlying mechanisms of chronic RIC and physical exercise in protecting multiple organs and benefiting various populations, the advantages of chronic RIC, and the challenges for its popularization. Intriguingly, several underlying mechanisms of RIC and exercise have been shown to overlap. These include the production of many autacoids, enhanced ability for antioxidant activity, modulating immune and inflammatory responses. Therefore, it appears that chronic RIC, just like regular exercise, has beneficial effects in unhealthy, sub-healthy and healthy individuals. Compared with regular exercise, chronic RIC has several advantages, which may provide novel insights into the area of exercise and health. Chronic RIC may enrich the modes of exercise, and benefit individuals with severe diseases. Also, the disabled, and sub-healthy individuals are likely to benefit from chronic RIC either as an alternative to exercise or an adjunct to pharmacological or non-pharmacological therapy.
Collapse
Affiliation(s)
- Wenbo Zhao
- ¹Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,2Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Sijie Li
- 2Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,3Beijing Municipal Geriatric Medical Research Center, Beijing, China.,4National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Changhong Ren
- 2Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,3Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Ran Meng
- ¹Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- 2Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,4National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
39
|
Yan BC, Wang J, Cao J, Won MH. Less hippocampal neuronal death in young gerbils following transient global cerebral ischemia is associated with long‑term maintenance of insulin‑like growth factor 1 and its receptors in the hippocampal CA1 region. Mol Med Rep 2017; 17:3055-3061. [PMID: 29257289 PMCID: PMC5783526 DOI: 10.3892/mmr.2017.8243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/13/2017] [Indexed: 01/03/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a well-known growth factor with well-defined neuroprotective effects against cerebral ischemia. However, the age‑dependent differences in the expression of IGF‑1 and its receptor (IGF‑1R) in the brain following transient cerebral ischemia (TCI) have not been elucidated. In the present study, the differences in IGF‑1 and IGF‑1R in the gerbil hippocampal CA1 region of young and adult gerbils 5 min following TCI were determined. Seven days following TCI, the neuronal death in the hippocampal CA1 region of young gerbils was significantly less than that observed in adult gerbils. In addition, the immunoreactivity, and levels of IGF‑1 and IGF‑1R in the CA1 region of the normal young were higher than those in the normal adult. Four days following TCI, the immunoreactivity, and protein levels of IGF‑1 and IGF‑1R were markedly decreased in the adult group. By contrast, in the young group, the immunoreactivity and expression levels were much greater than those in the adult group. However, 7 days following TCI, all immunoreactivity and expression levels were markedly decreased when compared with those in the normal adult and young groups. In addition, the immunoreactivity and expression levels in the young groups were significantly higher than those of the adult groups. In conclusion, the present study demonstrated that the higher and sustained expression of IGF‑1 and IGF‑1R in the young gerbil hippocampal CA1 region following TCI may be associated with the reduced neuronal death compared to that in the adults.
Collapse
Affiliation(s)
- Bing Chun Yan
- Department of Traditional Chinese and Western Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| | - Jie Wang
- Department of Traditional Chinese and Western Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| | - Jianwen Cao
- Department of Traditional Chinese and Western Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| |
Collapse
|