1
|
Wu G, Li B, Wei X, Chen Y, Zhao Y, Peng Y, Su J, Hu Z, Zhuo L, Tian Y, Wang Z, Peng X. Design, synthesis and biological evaluation of N-salicyloyl tryptamine derivatives as multifunctional neuroprotectants for the treatment of ischemic stroke. Eur J Med Chem 2024; 278:116795. [PMID: 39216381 DOI: 10.1016/j.ejmech.2024.116795] [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: 03/28/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Ischemic stroke (IS) is a disease of high death and disability worldwide with few medications in clinical treatment. Neuroinflammation and oxidative stress are considered as crucial factors in the progression of IS. In our previous studies, N-salicyloyl tryptamine derivative (NST) L7 exhibited promising anti-inflammatory properties and is considered a potential clinical therapy for IS but had limited antioxidant capacity. Here, we have designed, synthesized, and biologically evaluated 30 novel NSTs for their neuroprotective effects against cerebral ischemia-reperfusion (CI/R) injury. To identify a multifunctional neuroprotectant with enhanced antioxidant and anti-inflammatory capacity, as well as an effective therapeutic agent for CI/R damage. Among them, M11 exhibited synergistic highly anti-oxidant, anti-inflammatory, anti-ferroptosis, and anti-apoptosis effects and surpassed the parent compound L7. Further studies demonstrated that the synergistic and efficient neuroprotective role of M11 was mainly achieved by activating Nrf2 and stimulating its downstream target HO-1/GCLC/NQO1/GPX4. In addition, M11 possessed good blood-brain barrier permeability. Moreover, M11 effectively reduced cerebral infarct volume and improved neurological deficits in MCAO/R mice. Its hydrochloride form, M11·HCl, exhibited better pharmacokinetic properties, high safety, and a significant reduction in infarct volume, which is comparable to Edaravone. In conclusion, our findings suggested that M11 capable of activating Nrf2, could represent a promising candidate agent for IS.
Collapse
Affiliation(s)
- Genping Wu
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Bo Li
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiuzhen Wei
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yaxin Chen
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yuting Zhao
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yan Peng
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jianhui Su
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zecheng Hu
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Linsheng Zhuo
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ying Tian
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Wang
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China; MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, China.
| | - Xue Peng
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
2
|
Li J, Long Q, Ding H, Wang Y, Luo D, Li Z, Zhang W. Progress in the Treatment of Central Nervous System Diseases Based on Nanosized Traditional Chinese Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308677. [PMID: 38419366 PMCID: PMC11040388 DOI: 10.1002/advs.202308677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/07/2024] [Indexed: 03/02/2024]
Abstract
Traditional Chinese Medicine (TCM) is widely used in clinical practice to treat diseases related to central nervous system (CNS) damage. However, the blood-brain barrier (BBB) constitutes a significant impediment to the effective delivery of TCM, thus substantially diminishing its efficacy. Advances in nanotechnology and its applications in TCM (also known as nano-TCM) can deliver active ingredients or components of TCM across the BBB to the targeted brain region. This review provides an overview of the physiological and pathological mechanisms of the BBB and systematically classifies the common TCM used to treat CNS diseases and types of nanocarriers that effectively deliver TCM to the brain. Additionally, drug delivery strategies for nano-TCMs that utilize in vivo physiological properties or in vitro devices to bypass or cross the BBB are discussed. This review further focuses on the application of nano-TCMs in the treatment of various CNS diseases. Finally, this article anticipates a design strategy for nano-TCMs with higher delivery efficiency and probes their application potential in treating a wider range of CNS diseases.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
| | - Qingyin Long
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
| | - Huang Ding
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
| | - Yang Wang
- Institute of Integrative MedicineDepartment of Integrated Traditional Chinese and Western MedicineXiangya HospitalCentral South University ChangshaChangsha410008China
| | - Dan Luo
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
| | - Zhou Li
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
| | - Wei Zhang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
| |
Collapse
|
3
|
Zhang L, Han Y, Wu X, Chen B, Liu S, Huang J, Kong L, Wang G, Ye Z. Research progress on the mechanism of curcumin in cerebral ischemia/reperfusion injury: a narrative review. Apoptosis 2023; 28:1285-1303. [PMID: 37358747 DOI: 10.1007/s10495-023-01869-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2023] [Indexed: 06/27/2023]
Abstract
Cerebral ischemia/reperfusion (I/R) injury can result in different levels of cerebral impairment, and in severe cases, death. Curcumin, an essential bioactive component of turmeric, has a rich history as a traditional medicine for various ailments in numerous countries. Experimental and clinical research has established that curcumin offers a protective effect against cerebral I/R injury. Curcumin exerts its protective effects by acting on specific mechanisms such as antioxidant, anti-inflammatory, inhibition of ferroptosis and pyroptosis, protection of mitochondrial function and structure, reduction of excessive autophagy, and improvement of endoplasmic reticulum (ER) stress, which ultimately help to preserve the blood-brain barrier (BBB) and reducing apoptosis. There is currently a shortage of drugs undergoing clinical trials for the treatment of cerebral I/R injury, highlighting the pressing need for research and development of novel treatments to address this injury. The primary objective of this study is to establish a theoretical basis for future clinical applications of curcumin by delineating the mechanisms and protective effects of curcumin against cerebral I/R injury. Adapted with permission from [1].
Collapse
Affiliation(s)
- Liyuan Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Yibo Han
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Xuelan Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Baoyu Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Shuaiyuan Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Junyang Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Central Hospital of Chongqing University, Chongqing Emergency Medical Center, Chongqing, 400014, People's Republic of China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Zhiyi Ye
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
- JinFeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
4
|
Liu Y, Chen S, Liu S, Wallace KL, Zille M, Zhang J, Wang J, Jiang C. T-cell receptor signaling modulated by the co-receptors: Potential targets for stroke treatment. Pharmacol Res 2023; 192:106797. [PMID: 37211238 DOI: 10.1016/j.phrs.2023.106797] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/02/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Stroke is a severe and life-threatening disease, necessitating more research on new treatment strategies. Infiltrated T lymphocytes, an essential adaptive immune cell with extensive effector function, are crucially involved in post-stroke inflammation. Immediately after the initiation of the innate immune response triggered by microglia/macrophages, the adaptive immune response associated with T lymphocytes also participates in the complex pathophysiology of stroke and partially informs the outcome of stroke. Preclinical and clinical studies have revealed the conflicting roles of T cells in post-stroke inflammation and as potential therapeutic targets. Therefore, exploring the mechanisms that underlie the adaptive immune response associated with T lymphocytes in stroke is essential. The T-cell receptor (TCR) and its downstream signaling regulate T lymphocyte differentiation and activation. This review comprehensively summarizes the various molecules that regulate TCR signaling and the T-cell response. It covers both the co-stimulatory and co-inhibitory molecules and their roles in stroke. Because immunoregulatory therapies targeting TCR and its mediators have achieved great success in some proliferative diseases, this article also summarizes the advances in therapeutic strategies related to TCR signaling in lymphocytes after stroke, which can facilitate translation. DATA AVAILABILITY: No data was used for the research described in the article.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Shuai Chen
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Simon Liu
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, MD, 20814, USA
| | - Kevin L Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, A-1090 Vienna, Austria
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, 450000, Zhengzhou, P. R. China.
| | - Jian Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China; Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, P. R. China.
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China.
| |
Collapse
|
5
|
Yang X, Xu L, Zhao H, Xie T, Wang J, Wang L, Yang J. Curcumin protects against cerebral ischemia-reperfusion injury in rats by attenuating oxidative stress and inflammation: a meta-analysis and mechanism exploration. Nutr Res 2023; 113:14-28. [PMID: 36996692 DOI: 10.1016/j.nutres.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/11/2023] [Accepted: 02/26/2023] [Indexed: 03/09/2023]
Abstract
Accumulating evidence has suggested that curcumin may protect against cerebral ischemia-reperfusion injury (CIRI). However, biological mechanisms vary across studies, limiting the clinical applicability of these findings. We performed a meta-analysis on publications evaluating curcumin administration in rat models of CIRI. Furthermore, we sought to test the hypothesis that curcumin alleviates CIRI through diminishing oxidation and inflammation. We searched PubMed, Embase, Web of Science, and Cochrane from the starting date of each database to May 2022 for experimental rat studies exploring the use of curcumin after ischemia reperfusion. Included articles were assessed for bias using SYRCLE's risk of bias tool. Data were aggregated by a random effects model. Curcumin administration significantly reduced neurological deficit score (20 studies; pooled mean difference [MD] = -1.57; 95% CI, -1.78 to -1.36, P < .00001), infarct volume (18 studies; pooled MD = -17.56%; 95% CI, -20.92% to -14.20%; P < 0.00001), and brain water content (8 studies, pooled MD = -11.29%, 95% CI: -16.48%, -6.11%, P < .00001). Compared with control, the levels of superoxide dismutase, glutathione, and glutathione peroxidase were significantly higher, whereas the levels of reactive oxygen species, malondialdehyde, interleukin-1β, interleukin-6, interleukin-8, and nuclear factor kappa B were significantly lower (P < .05). Subgroup analysis raised the possibility that intervention affections differed by curcumin's dose. To our knowledge, this is the first meta-analysis of curcumin's neuroprotection and mechanisms in rat CIRI models. Our analysis suggests the neuroprotective potential of curcumin in CIRI via antioxidant activity and anti-inflammatory effect. More research is required to further confirm the effectiveness and safety of curcumin on ischemic stroke therapy.
Collapse
Affiliation(s)
- Xuyi Yang
- School of Agriculture and Bioengineering, Taizhou Vocational College of Science and Technology, Taizhou, China
| | - Liang Xu
- School of Agriculture and Bioengineering, Taizhou Vocational College of Science and Technology, Taizhou, China
| | - Hui Zhao
- Department of Critical Care Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Tinghui Xie
- School of Agriculture and Bioengineering, Taizhou Vocational College of Science and Technology, Taizhou, China
| | - Jiabing Wang
- Department of Pharmacy, Taizhou Municipal Hospital, Taizhou, China
| | - Lei Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianwei Yang
- General Practice, Zhejiang Taizhou Hospital, Linhai, China.
| |
Collapse
|
6
|
Polyphenols for the Treatment of Ischemic Stroke: New Applications and Insights. Molecules 2022; 27:molecules27134181. [PMID: 35807426 PMCID: PMC9268254 DOI: 10.3390/molecules27134181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Ischemic stroke (IS) is a leading cause of death and disability worldwide. Currently, the main therapeutic strategy involves the use of intravenous thrombolysis to restore cerebral blood flow to prevent the transition of the penumbra to the infarct core. However, due to various limitations and complications, including the narrow time window in which this approach is effective, less than 10% of patients benefit from such therapy. Thus, there is an urgent need for alternative therapeutic strategies, with neuroprotection against the ischemic cascade response after IS being one of the most promising options. In the past few decades, polyphenolic compounds have shown great potential in animal models of IS because of their high biocompatibility and ability to target multiple ischemic cascade signaling pathways, although low bioavailability is an issue that limits the applications of several polyphenols. Here, we review the pathophysiological changes following cerebral ischemia and summarize the research progress regarding the applications of polyphenolic compounds in the treatment of IS over the past 5 years. Furthermore, we discuss several potential strategies for improving the bioavailability of polyphenolic compounds as well as some essential issues that remain to be addressed for the translation of the related therapies to the clinic.
Collapse
|
7
|
Zhang J, Liu S, Jiang L, Hou J, Yang Z. Curcumin Improves Cardiopulmonary Resuscitation Outcomes by Modulating Mitochondrial Metabolism and Apoptosis in a Rat Model of Cardiac Arrest. Front Cardiovasc Med 2022; 9:908755. [PMID: 35665263 PMCID: PMC9160380 DOI: 10.3389/fcvm.2022.908755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/05/2022] [Indexed: 11/22/2022] Open
Abstract
Background Curcumin, a diarylheptanoid chemical compound extracted from curcuma longa, exerts a variety of biological and pharmacological effects in numerous pathological conditions, including ischemia/reperfusion (I/R) injury. In this study, we investigated its role in post-resuscitation myocardial dysfunction in a rat model of cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) by targeting on mitochondrial metabolism and apoptosis. Methods Animals were randomized into three groups: sham, control and curcumin, with fifteen rats in each group. Ventricular fibrillation (VF) was induced in the rats of the control and curcumin groups. The rats in the two groups were untreated for 8 min, followed by CPR for 8 min. Placebo (saline) or curcumin was administered by intraperitoneal injection, respectively, 5 min after successful resuscitation. Myocardial function was measured at baseline and post-resuscitation for 6 h consecutively. Ten rats in each group were closely observed for an additional 66 h to analyze the survival status, and the remaining five were sacrificed for the measurement of mitochondrial parameters and cell apoptosis. Results Compared with the control group, myocardial function was significantly enhanced in the curcumin group, contributing to a better survival status. Curcumin treatment mitigated the depletion of superoxide dismutase (SOD) and the production of malondialdehyde (MDA). The structural damage of mitochondria was also alleviated, with improved conditions of mPTP and ΔΨm. Curcumin boosted the production of ATP and attenuated myocardial apoptosis. Cytochrome C, caspase-3 and its cleavage were suppressed by curcumin. Proteins closely related to the functional performance of mitochondria, including uncoupling protein 2 (UCP2) and uncoupling protein 3 (UCP3) were downregulated, while mitochondrial transcription factor A (mtTFA) was upregulated. Conclusion Curcumin improves the outcomes of CPR via alleviating myocardial dysfunction induced by I/R injury. It exhibits anti-oxidation properties. Moreover, it is capable of ameliorating mitochondrial structure and energy metabolism, as well as inhibiting the mitochondrial apoptosis pathway. UCP2, UCP3, and mtTFA might also be involved in curcumin mediated protective effects on mitochondria.
Collapse
|
8
|
Fan F, Lei M. Mechanisms Underlying Curcumin-Induced Neuroprotection in Cerebral Ischemia. Front Pharmacol 2022; 13:893118. [PMID: 35559238 PMCID: PMC9090137 DOI: 10.3389/fphar.2022.893118] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 03/28/2022] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is the leading cause of death and disability worldwide, and restoring the blood flow to ischemic brain tissues is currently the main therapeutic strategy. However, reperfusion after brain ischemia leads to excessive reactive oxygen species production, inflammatory cell recruitment, the release of inflammatory mediators, cell death, mitochondrial dysfunction, endoplasmic reticulum stress, and blood-brain barrier damage; these pathological mechanisms will further aggravate brain tissue injury, ultimately affecting the recovery of neurological functions. It has attracted the attention of researchers to develop drugs with multitarget intervention effects for individuals with cerebral ischemia. A large number of studies have established that curcumin plays a significant neuroprotective role in cerebral ischemia via various mechanisms, including antioxidation, anti-inflammation, anti-apoptosis, protection of the blood-brain barrier, and restoration of mitochondrial function and structure, restoring cerebral circulation, reducing infarct volume, improving brain edema, promoting blood-brain barrier repair, and improving the neurological functions. Therefore, summarizing the results from the latest literature and identifying the potential mechanisms of action of curcumin in cerebral ischemia will serve as a basis and guidance for the clinical applications of curcumin in the future.
Collapse
Affiliation(s)
- Feng Fan
- Department of Interventional Neuroradiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Lei
- Department of Neurology, The Third People’s Hospital of Henan Province, Zhengzhou, China
| |
Collapse
|
9
|
Liu W, Jiang P, Qiu L. Blocking of Caveolin-1 Attenuates Morphine-Induced Inflammation, Hyperalgesia, and Analgesic Tolerance via Inhibiting NLRP3 Inflammasome and ERK/c-JUN Pathway. J Mol Neurosci 2022; 72:1047-1057. [PMID: 35262905 DOI: 10.1007/s12031-022-01989-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/23/2022] [Indexed: 12/21/2022]
Abstract
Morphine is generally used to treat chronic pain in clinic. But long-term use of morphine can inevitably induce analgesic tolerance and hyperalgesia. Caveolin-1 is reported to affect morphine-mediated signaling transduction. However, the action mechanism of morphine-induced analgesic tolerance is still unknown. In this study, morphine-induced analgesic tolerance model was established in Sprague-Dawley rats. The effects of Caveolin-1 blocking on neuroinflammation and ERK/c-JUN pathway were then explored. Morphine can remarkably elevate the expression level of Caveolin-1. Based on paw withdrawal latency behavior test, we found that Caveolin-1 blocking can effectively attenuate morphine-induced analgesic tolerance and neuroinflammation. Activation of ERK/c-JUN significantly reversed the above influences caused by Caveolin-1 blocking. Taken together, blocking of Caveolin-1 can attenuate morphine-induced inflammation and analgesic tolerance through inhibiting NLRP3 inflammasome and ERK/c-JUN pathway.
Collapse
Affiliation(s)
- Wenling Liu
- Department of Anestyesiology, HuiZhou Municipal Central Hospital, No. 41, Eling North Road, Huizhou City, Guangdong Province, 516001, China
| | - Peng Jiang
- Department of Anestyesiology, HuiZhou Municipal Central Hospital, No. 41, Eling North Road, Huizhou City, Guangdong Province, 516001, China
| | - Liuji Qiu
- Department of Anestyesiology, HuiZhou Municipal Central Hospital, No. 41, Eling North Road, Huizhou City, Guangdong Province, 516001, China.
| |
Collapse
|
10
|
Targeting AMPK signaling in ischemic/reperfusion injury: From molecular mechanism to pharmacological interventions. Cell Signal 2022; 94:110323. [DOI: 10.1016/j.cellsig.2022.110323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/16/2022]
|
11
|
Yan L, Guo MS, Zhang Y, Yu L, Wu JM, Tang Y, Ai W, Zhu FD, Law BYK, Chen Q, Yu CL, Wong VKW, Li H, Li M, Zhou XG, Qin DL, Wu AG. Dietary Plant Polyphenols as the Potential Drugs in Neurodegenerative Diseases: Current Evidence, Advances, and Opportunities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5288698. [PMID: 35237381 PMCID: PMC8885204 DOI: 10.1155/2022/5288698] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/10/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), are characterized by the progressive degeneration of neurons. Although the etiology and pathogenesis of neurodegenerative diseases have been studied intensively, the mechanism is still in its infancy. In general, most neurodegenerative diseases share common molecular mechanisms, and multiple risks interact and promote the pathologic process of neurogenerative diseases. At present, most of the approved drugs only alleviate the clinical symptoms but fail to cure neurodegenerative diseases. Numerous studies indicate that dietary plant polyphenols are safe and exhibit potent neuroprotective effects in various neurodegenerative diseases. However, low bioavailability is the biggest obstacle for polyphenol that largely limits its adoption from evidence into clinical practice. In this review, we summarized the widely recognized mechanisms associated with neurodegenerative diseases, such as misfolded proteins, mitochondrial dysfunction, oxidative damage, and neuroinflammatory responses. In addition, we summarized the research advances about the neuroprotective effect of the most widely reported dietary plant polyphenols. Moreover, we discussed the current clinical study and application of polyphenols and the factors that result in low bioavailability, such as poor stability and low permeability across the blood-brain barrier (BBB). In the future, the improvement of absorption and stability, modification of structure and formulation, and the combination therapy will provide more opportunities from the laboratory into the clinic for polyphenols. Lastly, we hope that the present review will encourage further researches on natural dietary polyphenols in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lu Yan
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Min-Song Guo
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yue Zhang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Wei Ai
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Qi Chen
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- Department of Nursing, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Vincent Kam-Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Hua Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Mao Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Da-Lian Qin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
12
|
Jiao Y, Wang J, Xue M. Effect of remote limb ischemic post‐conditioning on the expression of miR‐21‐5p/PirB in the brain of rats with focal cerebral ischemia. Eur J Neurosci 2022; 55:1105-1117. [PMID: 35060207 DOI: 10.1111/ejn.15600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Yiming Jiao
- Department of Cerebrovascular Diseases The Second Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Jinlan Wang
- Department of Cerebrovascular Diseases The Second Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases The Second Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| |
Collapse
|
13
|
Appunni S, Gupta D, Rubens M, Ramamoorthy V, Singh HN, Swarup V. Deregulated Protein Kinases: Friend and Foe in Ischemic Stroke. Mol Neurobiol 2021; 58:6471-6489. [PMID: 34549335 DOI: 10.1007/s12035-021-02563-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/10/2021] [Indexed: 12/20/2022]
Abstract
Ischemic stroke is the third leading cause of mortality worldwide, but its medical management is still limited to the use of thrombolytics as a lifesaving option. Multiple molecular deregulations of the protein kinase family occur during the period of ischemia/reperfusion. However, experimental studies have shown that alterations in the expression of essential protein kinases and their pharmacological modulation can modify the neuropathological milieu and hasten neurophysiological recovery. This review highlights the role of key protein kinase members and their implications in the evolution of stroke pathophysiology. Activation of ROCK-, MAPK-, and GSK-3β-mediated pathways following neuronal ischemia/reperfusion injury in experimental conditions aggravate the neuropathology and delays recovery. Targeting ROCK, MAPK, and GSK-3β will potentially enhance myelin regeneration, improve blood-brain barrier (BBB) function, and suppress inflammation, which ameliorates neuronal survival. Conversely, protein kinases such as PKA, Akt, PKCα, PKCε, Trk, and PERK salvage neurons post-ischemia by mechanisms including enhanced toxin metabolism, restoring BBB integrity, neurotrophic effects, and apoptosis suppression. Certain protein kinases such as ERK1/2, JNK, and AMPK have favourable and unfavourable effects in salvaging ischemia-injured neurons. Targeting multiple protein kinase-mediated pathways simultaneously may improve neuronal recovery post-ischemia.
Collapse
Affiliation(s)
- Sandeep Appunni
- Department of Biochemistry, Government Medical College, Kozhikode, Kerala, India
| | - Deepika Gupta
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | | | | | - Himanshu Narayan Singh
- Department of Systems Biology, Columbia University Irving Medical Centre, New York City, NY, USA.
| | - Vishnu Swarup
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|