1
|
Yu J, Li X, Zhou M, Lu M, Ruan Z, Zou W, Yu S. Schisandrin B Inhibits LPS-Induced Endometritis Through Attenuating Ferroptosis via AMPK/PGC1α/Nrf2 Signalling Pathway. J Cell Mol Med 2024; 28:e70281. [PMID: 39654025 PMCID: PMC11628352 DOI: 10.1111/jcmm.70281] [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: 04/16/2024] [Revised: 11/05/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Endometritis is one of the common reproductive diseases in human and animal. In recent years, a number of studies have found that Schisandra B (Sch B), as a natural Chinese medicine extract, has antioxidant, anti-inflammatory and other biological activities. Based on the above, in this study, mice were used to conduct an in vivo experiment to investigate the effect and mechanism of Sch B on lipopolysaccharide (LPS)-induced endometritis. Haematoxylin and eosin (H&E) staining was used to detect the pathological changes of uterine tissue and western blot was used to detect the expression levels of signalling pathways and key genes for ferroptosis. The results showed that Sch B significantly inhibited the pathological injury of uterine tissue, myeloperoxidase (MPO) activity, the activation of NF-κB pathway and the production of TNF-α and IL-1β. Furthermore, Sch B effectively inhibited ferroptosis by inhibiting malondialdehyde (MDA) and iron production and promoting the expression of ferroptosis suppressor genes GPX4 and ferritin. In conclusion, Sch B inhibited LPS-induced endometritis through alleviating inflammatory response and ferroptosis via AMPK/PGC1α/Nrf2 signalling pathway.
Collapse
Affiliation(s)
- Jing Yu
- Department of EndocrinologyAffiliated Hospital of Changchun University of Traditional Chinese MedicineChangchunJilinChina
| | - Xuewei Li
- Department of GynaecologyAffiliated Hospital of Changchun University of Traditional Chinese MedicineChangchunJilinChina
| | - Min Zhou
- Department of GynaecologyAffiliated Hospital of Changchun University of Traditional Chinese MedicineChangchunJilinChina
| | - Min Lu
- Department of GynaecologyAffiliated Hospital of Changchun University of Traditional Chinese MedicineChangchunJilinChina
| | - Zheng Ruan
- Department of Traditional Chinese Medicine964th HospitalChangchunChina
| | - Wenshuang Zou
- Department of GastroenterologyThe Affiliated Hospital to Changchun University of Chinese MedicineChangchunJilinPeople's Republic of China
| | - Shaohui Yu
- Department of GynaecologyAffiliated Hospital of Changchun University of Traditional Chinese MedicineChangchunJilinChina
| |
Collapse
|
2
|
Zeng ZJ, Lin X, Yang L, Li Y, Gao W. Activation of Inflammasomes and Relevant Modulators for the Treatment of Microglia-mediated Neuroinflammation in Ischemic Stroke. Mol Neurobiol 2024; 61:10792-10804. [PMID: 38789893 DOI: 10.1007/s12035-024-04225-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024]
Abstract
As the brain's resident immune patrol, microglia mediate endogenous immune responses to central nervous system injury in ischemic stroke, thereby eliciting either neuroprotective or neurotoxic effects. The association of microglia-mediated neuroinflammation with the progression of ischemic stroke is evident through diverse signaling pathways, notably involving inflammasomes. Within microglia, inflammasomes play a pivotal role in promoting the maturation of interleukin-1β (IL-1β) and interleukin-18 (IL-18), facilitating pyroptosis, and triggering immune infiltration, ultimately leading to neuronal cell dysfunction. Addressing the persistent and widespread inflammation holds promise as a breakthrough in enhancing the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ze-Jie Zeng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaobing Lin
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Liu Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Wen Gao
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
3
|
Hosseini A, Sheibani M, Valipour M. Exploring the Therapeutic Potential of BBB-Penetrating Phytochemicals With p38 MAPK Modulatory Activity in Addressing Oxidative Stress-Induced Neurodegenerative Disorders, With a Focus on Alzheimer's Disease. Phytother Res 2024; 38:5598-5625. [PMID: 39300812 DOI: 10.1002/ptr.8329] [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: 02/12/2024] [Revised: 07/17/2024] [Accepted: 08/17/2024] [Indexed: 09/22/2024]
Abstract
Oxidative stress plays an important role in the occurrence of neurodegenerative diseases. Previous studies indicate a strong connection between oxidative stress, inappropriate activation of the p38 MAPK signaling pathway, and the pathogenesis of neurodegenerative diseases. Although antioxidant therapy is a valid strategy to alleviate these problems, the most important limitation of this approach is the ineffectiveness of drug administration due to the limited permeability of the BBB. Therefore, BBB-penetrating p38 MAPK modulators with proper antioxidant capacity could be useful in preventing/reducing the complications of neurodegenerative disorders. The current manuscript aims to review the therapeutic capabilities of some recently reviewed naturally occurring p38 MAPK inhibitors in the management of neurodegenerative problems such as Alzheimer's disease. In data collection, we tried to use more recent studies published in high-quality journals indexed in databases Scopus, Web of Science, PubMed, and so on, but no specific time frame was considered due to the nature of the study. Our evaluations indicate that natural compounds tanshinones, protoberberines, pinocembrin, osthole, rhynchophylline, oxymatrine, schisandrin, piperine, paeonol, ferulic acid, 6-gingerol, obovatol, and trolox have significant potential for use as supplements/adjuvants in the reduction of neurodegenerative-related problems. Our findings emphasize the usefulness of BBB-penetrating phytochemicals with p38 MAPK modulatory activity as potential therapeutic options against neurodegenerative disorders. Of course, the proper use of these compounds depends on considering their toxicity/safety profile and pharmacokinetic characteristics as well as the clinical conditions of users.
Collapse
Affiliation(s)
- Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sheibani
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Valipour
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Zhang L, Xue S, Fei C, Yu C, Li J, Li Y, Wang N, Chu F, Pan L, Duan X, Peng D. Protective effect of Tao Hong Si Wu Decoction against inflammatory injury caused by intestinal flora disorders in an ischemic stroke mouse model. BMC Complement Med Ther 2024; 24:124. [PMID: 38500092 PMCID: PMC10946105 DOI: 10.1186/s12906-024-04417-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/27/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND AND AIMS Recent studies have shown that intestinal flora are involved in the pathological process of ischemic stroke (IS). The potential protective effect of the traditional Chinese prescription, Tao Hong Si Wu Decoction (THSWD), against inflammatory injury after IS and its underlying mechanisms of action were investigated in the current study. METHODS Fifty SPF(Specefic pathogen Free) male C57 mice were randomly assigned to sham operation, model, THSWD low-dose (6.5 g/kg), medium-dose (13 g/kg) and high-dose (26 g/kg) groups (10 mice per group). Mouse models of transient middle cerebral artery occlusion were prepared via thread embolism. Neurological function score, hematoxylin-eosin (HE) staining, immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), 16S ribosomal DNA (rDNA) sequencing, quantitative reverse transcription PCR (qRT-PCR) and other methods were employed to elucidate the underlying molecular mechanisms. RESULTS Notably, THSWD induced a reduction in the neurological function score (P < 0.01) and neuronal injury in brain tissue, increase in protein expression of Claudin-5 and zonula occludens-1 (ZO-1) in brain tissue(P < 0.01), and decrease in serum lipopolysaccharide (LPS)(P < 0.01), diamine oxidase (DAO)(P < 0.01) and D-lactic acid(P < 0.01, P < 0.05) levels to a significant extent. THSWD also inhibited the levels of tumor necrosis factor-α (TNF-α)(P < 0.01) and interleukin - 1β (IL-1β)(P < 0.01) in brain tissue, and increased alpha and beta diversity in ischemic stroke mice, along with a certain reversal effect on different microflora. Finally, THSWD inhibited the polarization of microglia cells(P < 0.01) and decreased the protein and gene expression of toll-like receptor-4 (TLR-4)(P < 0.01, P < 0.05) and nuclear factor kappa B (NF-κB)(P < 0.01) in brain tissue. CONCLUSION Our data indicate that THSWD may interfere with inflammatory response in ischemic stroke by regulating intestinal flora and promoting intestinal barrier repair.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Sujun Xue
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Changyi Fei
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Chao Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Jingjing Li
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yumeng Li
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ni Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Furui Chu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Lingyu Pan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China
| | - Xianchun Duan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, China.
- Key Laboratory of Chinese Medicinal Formula Research, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
- Key Laboratory of Chinese Medicinal Formula Research, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
5
|
Wróbel-Biedrawa D, Podolak I. Anti-Neuroinflammatory Effects of Adaptogens: A Mini-Review. Molecules 2024; 29:866. [PMID: 38398618 PMCID: PMC10891670 DOI: 10.3390/molecules29040866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Introduction: Adaptogens are a group of plants that exhibit complex, nonspecific effects on the human body, increasing its ability to adapt, develop resilience, and survive in stress conditions. They are found in many traditional medicinal systems and play a key role in restoring the body's strength and stamina. Research in recent years has attempted to elucidate the mechanisms behind their pharmacological effects, but it appears that these effects are difficult to define precisely and involve multiple molecular pathways. Neuroinflammation: In recent years, chronic inflammation has been recognized as one of the common features of many central nervous system disorders (dementia and other neurodegenerative diseases, depression, anxiety, ischemic stroke, and infections). Because of the specific nature of the brain, this process is called neuroinflammation, and its suppression can result in an improvement of patients' condition and may promote their recovery. Adaptogens as anti-inflammatory agents: As has been discovered, adaptogens display anti-inflammatory effects, which suggests that their application may be broader than previously thought. They regulate gene expression of anti- and proinflammatory cytokines (prostaglandins, leukotriens) and can modulate signaling pathways (e.g., NF-κB). Aim: This mini-review aims to present the anti-neuroinflammatory potential of the most important plants classified as adaptogens: Schisandra chinensis, Eleutherococcus senticosus, Rhodiola rosea and Withania somnifera.
Collapse
Affiliation(s)
| | - Irma Podolak
- Department of Pharmacognosy, Jagiellonian University Collegium Medicum, Medyczna 9, 30-688 Cracow, Poland;
| |
Collapse
|
6
|
Sharma V, Sharma P, Singh TG. Therapeutic Correlation of TLR-4 Mediated NF-κB Inflammatory Pathways in Ischemic Injuries. Curr Drug Targets 2024; 25:1027-1040. [PMID: 39279711 DOI: 10.2174/0113894501322228240830063605] [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: 04/09/2024] [Revised: 07/06/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024]
Abstract
Ischemia-reperfusion (I/R) injury refers to the tissue damage that happens when blood flow returns to tissue after a period of ischemia. I/R injuries are implicated in a large array of pathological conditions, such as cerebral, myocardial, renal, intestinal, retinal and hepatic ischemia. The hallmark of these pathologies is excessive inflammation. Toll-like receptors (TLRs) are recognized as significant contributors to inflammation caused by pathogens and, more recently, inflammation caused by injury. TLR-4 activation initiates a series of events that results in activation of nuclear factor kappa-B (NF-κB), which stimulates the production of pro-inflammatory cytokines and chemokines, exacerbating tissue injury. Therefore, through a comprehensive review of current research and experimentation, this investigation elucidates the TLRs signalling pathway and the role of TLR-4/NF-κB in the pathophysiology of I/R injuries. Furthermore, this review highlights the various pharmacological agents (TLR-4/NF-κB inhibitors) with special emphasis on the various ischemic injuries (cerebral, myocardial, renal, intestinal, retinal and hepatic). Future research should prioritise investigating the specific molecular pathways that cause TLR-4/NF-κBmediated inflammation in ischemic injuries. Additionally, efforts should be made to enhance treatment approaches in order to enhance patient outcomes.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Prateek Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
7
|
Chen X, Wang W, Li H, Zhang X. Enriched environment alleviates neurological deficits via downregulation of Cx43 after experimental stroke. Brain Res 2023; 1821:148619. [PMID: 37805009 DOI: 10.1016/j.brainres.2023.148619] [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: 06/08/2023] [Revised: 09/21/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
While it has been demonstrated that enriched environment (EE) can protect against cerebral ischemia/reperfusion (I/R) injury, the underlying mechanism remains largely unknown. Connexin 43 (Cx43) is a key component of gap junctions, which may mediate cell-to-cell communication in neural cells. This study aimed to investigate the neuroprotective effects of EE against cerebral I/R injury in rats by modulating Cx43. A rat model of cerebral I/R injury was established by middle cerebral artery occlusion (MCAO)/reperfusion. Rats were randomly divided into the sham, MCAO, MCAO + EE, MCAO + Gap19, and MCAO + EE + Gap19 groups. The modified neurological severity score test and Morris water maze assay were used to assess neurological deficits. The infarct volume was measured using triphenyltetrazolium chloride (TTC) staining. Neuronal survival was detected by immunofluorescence. The indices of oxidative stress were determined using ELISA, and the reactive oxygen species levels were determined using a dihydroethidium probe. Cx43 and inflammation-related protein expression levels were also measured using western blotting and immunohistochemistry. EE and Gap19 treatment significantly improved neurological deficits, reduced infarct volumes, attenuated neuronal injury, and suppressed inflammatory cytokine expression and oxidative stress. Furthermore, EE and Gap19 treatment notably downregulated the expression of Cx43 and the inflammation-related pathway TLR4/MyD88/NF-κB in the ischemic penumbra. Gap19, a Cx43 inhibitor, markedly enhanced the neuroprotective effects of EE in rats with cerebral I/R injury. EE treatment protects against cerebral I/R injury in rats via Cx43 downregulation. Our findings may shed light on the mechanism underlying the protective efficacy of EE.
Collapse
Affiliation(s)
- Xiuping Chen
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wansong Wang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hui Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Ruijin 2rd, Shanghai 200025, China.
| | - Xin Zhang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuhan 430071, Hubei, China.
| |
Collapse
|
8
|
Nieoczym D, Banono NS, Stępnik K, Kaczor AA, Szybkowski P, Esguerra CV, Kukula-Koch W, Gawel K. In Silico Analysis, Anticonvulsant Activity, and Toxicity Evaluation of Schisandrin B in Zebrafish Larvae and Mice. Int J Mol Sci 2023; 24:12949. [PMID: 37629132 PMCID: PMC10455331 DOI: 10.3390/ijms241612949] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The aim of this study is to evaluate the anticonvulsant potential of schisandrin B, a main ingredient of Schisandra chinensis extracts. Schisandrin B showed anticonvulsant activity in the zebrafish larva pentylenetetrazole acute seizure assay but did not alter seizure thresholds in the intravenous pentylenetetrazole test in mice. Schisandrin B crosses the blood-brain barrier, which we confirmed in our in silico and in vivo analyses; however, the low level of its unbound fraction in the mouse brain tissue may explain the observed lack of anticonvulsant activity. Molecular docking revealed that the anticonvulsant activity of the compound in larval zebrafish might have been due to its binding to a benzodiazepine site within the GABAA receptor and/or the inhibition of the glutamate NMDA receptor. Although schisandrin B showed a beneficial anticonvulsant effect, toxicological studies revealed that it caused serious developmental impairment in zebrafish larvae, underscoring its teratogenic properties. Further detailed studies are needed to precisely identify the properties, pharmacological effects, and safety of schisandrin B.
Collapse
Affiliation(s)
- Dorota Nieoczym
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Nancy Saana Banono
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, University of Oslo, Gaustadalleen 21, Forskningsparken, 0349 Oslo, Norway; (N.S.B.); (C.V.E.)
| | - Katarzyna Stępnik
- Department of Physical Chemistry, Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Skłodowska University, Pl. M. Curie-Skłodowskiej 3/243, 20-031 Lublin, Poland;
| | - Agnieszka A. Kaczor
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20-093 Lublin, Poland;
| | - Przemysław Szybkowski
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego St. 8b, 20-090 Lublin, Poland;
- Clinical Provincial Hospital No. 2 St. Jadwiga Krolowej in Rzeszow, Lwowska St. 60, 35-301 Rzeszow, Poland
| | - Camila Vicencio Esguerra
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, University of Oslo, Gaustadalleen 21, Forskningsparken, 0349 Oslo, Norway; (N.S.B.); (C.V.E.)
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, Chodźki St. 1, 20-093 Lublin, Poland;
| | - Kinga Gawel
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego St. 8b, 20-090 Lublin, Poland;
| |
Collapse
|
9
|
Zheng T, Jiang T, Huang Z, Ma H, Wang M. Role of traditional Chinese medicine monomers in cerebral ischemia/reperfusion injury:a review of the mechanism. Front Pharmacol 2023; 14:1220862. [PMID: 37654609 PMCID: PMC10467294 DOI: 10.3389/fphar.2023.1220862] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/04/2023] [Indexed: 09/02/2023] Open
Abstract
Ischemia/reperfusion (I/R) injury is a pathological process wherein reperfusion of an ischemic organ or tissue exacerbates the injury, posing a significant health threat and economic burden to patients and their families. I/R triggers a multitude of physiological and pathological events, such as inflammatory responses, oxidative stress, neuronal cell death, and disruption of the blood-brain barrier (BBB). Hence, the development of effective therapeutic strategies targeting the pathological processes resulting from I/R is crucial for the rehabilitation and long-term enhancement of the quality of life in patients with cerebral ischemia/reperfusion injury (CIRI). Traditional Chinese medicine (TCM) monomers refer to bioactive compounds extracted from Chinese herbal medicine, possessing anti-inflammatory and antioxidative effects, and the ability to modulate programmed cell death (PCD). TCM monomers have emerged as promising candidates for the treatment of CIRI and its subsequent complications. Preclinical studies have demonstrated that TCM monomers can enhance the recovery of neurological function following CIRI by mitigating oxidative stress, suppressing inflammatory responses, reducing neuronal cell death and functional impairment, as well as minimizing cerebral infarction volume. The neuroprotective effects of TCM monomers on CIRI have been extensively investigated, and a comprehensive understanding of their mechanisms can pave the way for novel approaches to I/R treatment. This review aims to update and summarize evidence of the protective effects of TCMs in CIRI, with a focus on their role in modulating oxidative stress, inflammation, PCD, glutamate excitotoxicity, Ca2+ overload, as well as promoting blood-brain barrier repairment and angiogenesis. The main objective is to underscore the significant contribution of TCM monomers in alleviating CIRI.
Collapse
Affiliation(s)
| | | | | | | | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
10
|
Ri MH, Xing Y, Zuo HX, Li MY, Jin HL, Ma J, Jin X. Regulatory mechanisms of natural compounds from traditional Chinese herbal medicines on the microglial response in ischemic stroke. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154889. [PMID: 37262999 DOI: 10.1016/j.phymed.2023.154889] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 04/12/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND Development of clinically effective neuroprotective agents for stroke therapy is still a challenging task. Microglia play a critical role in brain injury and recovery after ischemic stroke. Traditional Chinese herbal medicines (TCHMs) are based on a unique therapeutic principle, have various formulas, and have long been widely used to treat stroke. Therefore, the active compounds in TCHMs and their underlying mechanisms of action are attracting increasing attention in the field of stroke drug development. PURPOSE To summarize the regulatory mechanisms of TCHM-derived natural compounds on the microglial response in animal models of ischemic stroke. METHODS We searched studies published until 10 April 2023 in the Web of Science, PubMed, and ScienceDirect using the following keywords: natural compounds, natural products or phytochemicals, traditional Chinese Medicine or Chinese herbal medicine, microglia, and ischemic stroke. This review was prepared according to PRISMA (Preferred Reporting Item for Systematic Reviews and Meta-Analysis) guidelines. RESULTS Natural compounds derived from TCHMs can attenuate the M1 phenotype of microglia, which is involved in the detrimental inflammatory response, via inhibition of NF-κB, MAPKs, JAK/STAT, Notch, TLR4, P2X7R, CX3CR1, IL-17RA, the NLRP3 inflammasome, and pro-oxidant enzymes. Additionally, the neuroprotective response of microglia with the M2 phenotype can be enhanced by activating Nrf2/HO-1, PI3K/AKT, AMPK, PPARγ, SIRT1, CB2R, TREM2, nAChR, and IL-33/ST2. Several clinical trials showed that TCHM-derived natural compounds that regulate microglial responses have significant and safe therapeutic effects, but further well-designed clinical studies are needed. CONCLUSIONS Further research regarding the direct targets and potential pleiotropic or synergistic effects of natural compounds would provide a more reasonable approach for regulation of the microglial response with the possibility of successful stroke drug development.
Collapse
Affiliation(s)
- Myong Hak Ri
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China; Faculty of Life Science, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Yue Xing
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Hong Xiang Zuo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Ming Yue Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Hong Lan Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Juan Ma
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| | - Xuejun Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| |
Collapse
|
11
|
Lam HYP, Liang TR, Peng SY. Prevention of the Pro-Aggressive Effects of Ethanol-Intoxicated Mice by Schisandrin B. Nutrients 2023; 15:nu15081909. [PMID: 37111128 PMCID: PMC10146817 DOI: 10.3390/nu15081909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Excessive alcohol consumption can lead to serious health complications, with liver and neurological complications being the most important. In Western nations, alcoholic liver disease accounts for 50% of mortality from end-stage liver disease and is the second most common cause of liver transplants. In addition to direct damage, hepatic encephalopathy may also arise from alcohol consumption. However, effective treatment for liver disease, as well as neurological injury, is still lacking today; therefore, finding an efficacious alternative is urgently needed. In the current study, the preventive and therapeutic effects of Schisandrin B (Sch B) against ethanol-induced liver and brain injuries were investigated. By using two treatment models, our findings indicated that Sch B can effectively prevent and ameliorate alcoholic liver diseases, such as resolving liver injuries, lipid deposition, inflammasome activation, and fibrosis. Moreover, Sch B reverses brain damage and improves the neurological function of ethanol-treated mice. Therefore, Sch B may serve as a potential treatment option for liver diseases, as well as subsequential brain injuries. Furthermore, Sch B may be useful in preventive drug therapy against alcohol-related diseases.
Collapse
Affiliation(s)
- Ho Yin Pekkle Lam
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Ting-Ruei Liang
- PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien 970, Taiwan
| | - Shih-Yi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
12
|
Guo S, Wehbe A, Syed S, Wills M, Guan L, Lv S, Li F, Geng X, Ding Y. Cerebral Glucose Metabolism and Potential Effects on Endoplasmic Reticulum Stress in Stroke. Aging Dis 2023; 14:450-467. [PMID: 37008060 PMCID: PMC10017147 DOI: 10.14336/ad.2022.0905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
Ischemic stroke is an extremely common pathology with strikingly high morbidity and mortality rates. The endoplasmic reticulum (ER) is the primary organelle responsible for conducting protein synthesis and trafficking as well as preserving intracellular Ca2+ homeostasis. Mounting evidence shows that ER stress contributes to stroke pathophysiology. Moreover, insufficient circulation to the brain after stroke causes suppression of ATP production. Glucose metabolism disorder is an important pathological process after stroke. Here, we discuss the relationship between ER stress and stroke and treatment and intervention of ER stress after stroke. We also discuss the role of glucose metabolism, particularly glycolysis and gluconeogenesis, post-stroke. Based on recent studies, we speculate about the potential relationship and crosstalk between glucose metabolism and ER stress. In conclusion, we describe ER stress, glycolysis, and gluconeogenesis in the context of stroke and explore how the interplay between ER stress and glucose metabolism contributes to the pathophysiology of stroke.
Collapse
Affiliation(s)
- Sichao Guo
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Alexandra Wehbe
- Department of Neurosurgery, Wayne State University School of Medicine, USA
- Harvard T.H. Chan School of Public Health, USA
| | - Shabber Syed
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Melissa Wills
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Longfei Guan
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Shuyu Lv
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, USA
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| |
Collapse
|
13
|
Qiao C, Liu Z, Qie S. The Implications of Microglial Regulation in Neuroplasticity-Dependent Stroke Recovery. Biomolecules 2023; 13:biom13030571. [PMID: 36979506 PMCID: PMC10046452 DOI: 10.3390/biom13030571] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Stroke causes varying degrees of neurological deficits, leading to corresponding dysfunctions. There are different therapeutic principles for each stage of pathological development. Neuroprotection is the main treatment in the acute phase, and functional recovery becomes primary in the subacute and chronic phases. Neuroplasticity is considered the basis of functional restoration and neurological rehabilitation after stroke, including the remodeling of dendrites and dendritic spines, axonal sprouting, myelin regeneration, synapse shaping, and neurogenesis. Spatiotemporal development affects the spontaneous rewiring of neural circuits and brain networks. Microglia are resident immune cells in the brain that contribute to homeostasis under physiological conditions. Microglia are activated immediately after stroke, and phenotypic polarization changes and phagocytic function are crucial for regulating focal and global brain inflammation and neurological recovery. We have previously shown that the development of neuroplasticity is spatiotemporally consistent with microglial activation, suggesting that microglia may have a profound impact on neuroplasticity after stroke and may be a key therapeutic target for post-stroke rehabilitation. In this review, we explore the impact of neuroplasticity on post-stroke restoration as well as the functions and mechanisms of microglial activation, polarization, and phagocytosis. This is followed by a summary of microglia-targeted rehabilitative interventions that influence neuroplasticity and promote stroke recovery.
Collapse
Affiliation(s)
- Chenye Qiao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| |
Collapse
|
14
|
Zhao N, Gao Y, Jia H, Jiang X. Anti-apoptosis effect of traditional Chinese medicine in the treatment of cerebral ischemia-reperfusion injury. Apoptosis 2023; 28:702-729. [PMID: 36892639 DOI: 10.1007/s10495-023-01824-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2023] [Indexed: 03/10/2023]
Abstract
Cerebral ischemia, one of the leading causes of neurological dysfunction of brain cells, muscle dysfunction, and death, brings great harm and challenges to individual health, families, and society. Blood flow disruption causes decreased glucose and oxygen, insufficient to maintain normal brain tissue metabolism, resulting in intracellular calcium overload, oxidative stress, neurotoxicity of excitatory amino acids, and inflammation, ultimately leading to neuronal cell necrosis, apoptosis, or neurological abnormalities. This paper summarizes the specific mechanism of cell injury that apoptosis triggered by reperfusion after cerebral ischemia, the related proteins involved in apoptosis, and the experimental progress of herbal medicine treatment through searching, analyzing, and summarizing the PubMed and Web Of Science databases, which includes active ingredients of herbal medicine, prescriptions, Chinese patent medicines, and herbal extracts, providing a new target or new strategy for drug treatment, and providing a reference for future experimental directions and using them to develop suitable small molecule drugs for clinical application. With the research of anti-apoptosis as the core, it is important to find highly effective, low toxicity, safe and cheap compounds from natural plants and animals with abundant resources to prevent and treat Cerebral ischemia/reperfusion (I/R) injury (CIR) and solve human suffering. In addition, understanding and summarizing the apoptotic mechanism of cerebral ischemia-reperfusion injury, the microscopic mechanism of CIR treatment, and the cellular pathways involved will help to develop new drugs.
Collapse
Affiliation(s)
- Nan Zhao
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yuhe Gao
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Hongtao Jia
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xicheng Jiang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China.
| |
Collapse
|
15
|
Induced Inflammatory and Oxidative Markers in Cerebral Microvasculature by Mentally Depressive Stress. Mediators Inflamm 2023; 2023:4206316. [PMID: 36852396 PMCID: PMC9966573 DOI: 10.1155/2023/4206316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/25/2022] [Accepted: 11/24/2022] [Indexed: 02/20/2023] Open
Abstract
Background Cerebrovascular disease (CVD) is recognized as the leading cause of permanent disability worldwide. Depressive disorders are associated with increased incidence of CVD. The goal of this study was to establish a chronic restraint stress (CRS) model for mice and examine the effect of stress on cerebrovascular inflammation and oxidative stress responses. Methods A total of forty 6-week-old male C57BL/6J mice were randomly divided into the CRS and control groups. In the CRS group (n = 20), mice were placed in a well-ventilated Plexiglas tube for 6 hours per day for 28 consecutive days. On day 29, open field tests (OFT) and sucrose preference tests (SPT) were performed to assess depressive-like behaviors for the two groups (n = 10/group). Macrophage infiltration into the brain tissue upon stress was analyzed by measuring expression of macrophage marker (CD68) with immunofluorescence in both the CRS and control groups (n = 10/group). Cerebral microvasculature was isolated from the CRS and controls (n = 10/group). mRNA and protein expressions of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), vascular cell adhesion molecule-1 (VCAM-1), and macrophage chemoattractant protein-1 (MCP-1) in the brain vessels were measured by real-time PCR and Western blot (n = 10/group). Reactive oxygen species (ROS), hydrogen peroxide (H2O2), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activities were quantified by ELISA to study the oxidative profile of the brain vessels (n = 10/group). Additionally, mRNA and protein expressions of NOX subunits (gp91phox, p47phox, p67phox, and p22phox) in the cerebrovascular endothelium were analyzed by real-time PCR and Western blot (n = 10/group). Results CRS decreased the total distances (p < 0.05) and the time spent in the center zone in OFT (p < 0.001) and sucrose preference test ratio in SPT (p < 0.01). Positive ratio of CD68+ was increased with CRS in the entire region of the brain (p < 0.001), reflecting increased macrophage infiltration. CRS increased the expression of inflammatory factors and oxidative stress in the cerebral microvasculature, including TNF-α (p < 0.001), IL-1β (p < 0.05), IL-6 (p < 0.05), VCAM-1 (p < 0.01), MCP-1 (p < 0.01), ROS (p < 0.001), and H2O2 (p < 0.001). NADPH oxidase (NOX) was activated by CRS (p < 0.01), and mRNA and protein expressions of NOX subunits (gp91phox, p47phox, p67phox, and p22phox) in brain microvasculature were found to be increased. Conclusions To our knowledge, this is the first study to demonstrate that CRS induces depressive stress and causes inflammatory and oxidative stress responses in the brain microvasculature.
Collapse
|
16
|
Li L, Pan G, Fan R, Li D, Guo L, Ma L, Liang H, Qiu J. Luteolin alleviates inflammation and autophagy of hippocampus induced by cerebral ischemia/reperfusion by activating PPAR gamma in rats. BMC Complement Med Ther 2022; 22:176. [PMID: 35778706 PMCID: PMC9248165 DOI: 10.1186/s12906-022-03652-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/23/2022] [Indexed: 11/10/2022] Open
Abstract
Background Luteolin, a flavonoid compound with anti-inflammatory activity, has been reported to alleviate cerebral ischemia/reperfusion (I/R) injury. However, its potential mechanism remains unclear. Methods The binding activity of luteolin to peroxisome proliferator-activated receptor gamma (PPARγ) was calculated via molecular docking analysis. Rats were subjected to middle cerebral artery occlusion and reperfusion (MCAO/R). After reperfusion, vehicle, 25 mg/kg/d luteolin, 50 mg/kg/d luteolin, 10 mg/kg/d pioglitazone, 50 mg/kg/d luteolin combined with 10 mg/kg/d T0070907 (PPARγ inhibitor) were immediately orally treatment for 7 days. ELISA, TTC staining, H&E staining, immunohistochemistry, immunofluorescence and transmission electron microscope methods were performed to evaluate the inflammation and autophagy in damaged hippocampal region. The PPARγ, light chain 3 (LC3) B-II/LC3B-I and p-nuclear factor-κB (NF-κB) p65 proteins expression levels in damaged hippocampal region were analyzed. Results Luteolin showed good PPARγ activity according to docking score (score = − 8.2). Luteolin treatment downregulated the infarct area and the pro-inflammatory cytokines levels caused by MCAO/R injury. Moreover, luteolin administration ameliorated neuroinflammation and autophagy in damaged hippocampal region. Pioglitazone plays protective roles similar to luteolin. T0070907 concealed the neuroprotective roles of 50 mg/kg/d luteolin. Conclusions Luteolin exerts neuroprotective roles against inflammation and autophagy of hippocampus induced by cerebral I/R by activating PPARγ in rats. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03652-8.
Collapse
|
17
|
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: 0.7] [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
|
18
|
The Signaling Pathways and Targets of Natural Compounds from Traditional Chinese Medicine in Treating Ischemic Stroke. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103099. [PMID: 35630576 PMCID: PMC9148018 DOI: 10.3390/molecules27103099] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022]
Abstract
Ischemic stroke (IS) is a common neurological disorder associated with high disability rates and mortality rates. At present, recombinant tissue plasminogen activator (r-tPA) is the only US(FDA)-approved drug for IS. However, due to the narrow therapeutic window and risk of intracerebral hemorrhage, r-tPA is currently used in less than 5% of stroke patients. Natural compounds have been widely used in the treatment of IS in China and have a wide range of therapeutic effects on IS by regulating multiple targets and signaling pathways. The keywords "ischemia stroke, traditional Chinese Medicine, Chinese herbal medicine, natural compounds" were used to search the relevant literature in PubMed and other databases over the past five years. The results showed that JAK/STAT, NF-κB, MAPK, Notch, Nrf2, and PI3K/Akt are the key pathways, and SIRT1, MMP9, TLR4, HIF-α are the key targets for the natural compounds from traditional Chinese medicine in treating IS. This study aims to update and summarize the signaling pathways and targets of natural compounds in the treatment of IS, and provide a base of information for the future development of effective treatments for IS.
Collapse
|
19
|
Ma R, Zhan Y, Zhang Y, Wu L, Wang X, Guo M. Schisandrin B ameliorates non-alcoholic liver disease through anti-inflammation activation in diabetic mice. Drug Dev Res 2022; 83:735-744. [PMID: 34927282 PMCID: PMC9299884 DOI: 10.1002/ddr.21905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 11/14/2021] [Accepted: 12/01/2021] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic risk factor associated with non-alcoholic liver disease (NAFLD). Schisandrin B (Sch B) is a promising agent for NAFLD. However, the actions of Sch B on diabetes-associated NAFLD and the underlying mechanisms are not characterized. This study aimed to assess whether Sch B has beneficial effects on T2DM-associated NAFLD. Sch B (50 mg/kg, gavage) was administrated to C57BL/KSJ db/db mice for 2 weeks. Body weight, liver weight, blood glucose, and insulin resistance were measured. Serum lipid level and liver function were detected using the biochemistry analyzer. Quantitative Real-Time PCR assay was used to evaluate mRNA levers of lipid metabolism genes. Terminal-deoxynucleoitidyl Transferase Mediated Nick End Labeling (TUNEL) staining was performed to measure apoptosis in the liver. Pathological analysis and immunohistochemistry assessment were used to analyze hepatic steatosis and inflammatory infiltration. Sch B supplementation significantly decrease body weight, related liver weight, blood glucose, and serum insulin, and improved insulin resistance in db/db mice. Sch B obviously corrected NAFLD phenotypes including lipid deposition, steatohepatitis, and high levels of hepatic enzymes and serum lipid. In addition, mRNA levels of Sterol response element-bind protein 1c (SREBP-1c), fatty acid synthetase (Fasn), and acetyl-CoA carboxylase (ACC) were markedly downregulated by Sch B treatment. TUNEL-positive cells were also decreased by Sch B. Furthermore, Sch B inhibited the Kupffer cells, IL-1β, and TNF-α infiltration to the liver. Sch B ameliorated insulin resistance and lipid accumulation under high glucose conditions, which was partly associated with its inhibition of apoptosis and anti-inflammatory actions.
Collapse
Affiliation(s)
- Ruojia Ma
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| | - Yike Zhan
- Department of CardiologyJieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat‐sen University)JieyangChina
| | - Yamei Zhang
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| | - Liangan Wu
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| | - Xing Wang
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| | - Ming Guo
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| |
Collapse
|
20
|
Guo S, Mangal R, Dandu C, Geng X, Ding Y. Role of Forkhead Box Protein O1 (FoxO1) in Stroke: A Literature Review. Aging Dis 2022; 13:521-533. [PMID: 35371601 PMCID: PMC8947839 DOI: 10.14336/ad.2021.0826] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022] Open
Abstract
Stroke is one of the most prevalent causes of death around the world. When a stroke occurs, many cellular signaling cascades and regulators are activated, which results in severe cellular dysfunction and debilitating long-term disability. One crucial regulator of cell fate and function is mammalian Forkhead box protein O1 (FoxO1). Many studies have found FoxO1 to be implicated in many cellular processes, including regulating gluconeogenesis and glycogenolysis. During a stroke, modifications of FoxO1 have been linked to a variety of functions, such as inducing cell death and inflammation, inhibiting oxidative injury, affecting the blood brain barrier (BBB), and regulating hepatic gluconeogenesis. For these functions of FoxO1, different measures and treatments were applied to FoxO1 after ischemia. However, the subtle mechanisms of post-transcriptional modification and the role of FoxO1 are still elusive and even contradictory in the development of stroke. The determination of these mechanisms will lead to further enlightenment for FoxO1 signal transduction and the identification of targeted drugs. The regulation and function of FoxO1 may provide an important way for the prevention and treatment of diseases. Overall, the functions of FoxO1 are multifactorial, and this paper will summarize all of the significant pathways in which FoxO1 plays an important role during stroke damage and recovery.
Collapse
Affiliation(s)
- Sichao Guo
- 1Luhe Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ruchi Mangal
- 3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Chaitu Dandu
- 3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xiaokun Geng
- 1Luhe Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,2Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yuchuan Ding
- 3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
21
|
Neural stem cell therapy in conjunction with curcumin loaded in niosomal nanoparticles enhanced recovery from traumatic brain injury. Sci Rep 2022; 12:3572. [PMID: 35246564 PMCID: PMC8897489 DOI: 10.1038/s41598-022-07367-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/16/2022] [Indexed: 12/28/2022] Open
Abstract
Despite a great amount of effort, there is still a need for reliable treatments of traumatic brain injury (TBI). Recently, stem cell therapy has emerged as a new avenue to address neuronal regeneration after TBI. However, the environment of TBI lesions exerts negative effects on the stem cells efficacy. Therefore, to maximize the beneficial effects of stem cells in the course of TBI, we evaluated the effect of human neural stem/progenitor cells (hNS/PCs) and curcumin-loaded niosome nanoparticles (CM-NPs) on behavioral changes, brain edema, gliosis, and inflammatory responses in a rat model of TBI. After TBI, hNS/PCs were transplanted within the injury site and CM-NPs were orally administered for 10 days. Finally, the effect of combination therapy was compared to several control groups. Our results indicated a significant improvement of general locomotor activity in the hNS/PCs + CM-NPs treatment group compared to the control groups. We also observed a significant improvement in brain edema in the hNS/PCs + CM-NPs treatment group compared to the other groups. Furthermore, a significant decrease in astrogliosis was seen in the combined treatment group. Moreover, TLR4-, NF-κB-, and TNF-α- positive cells were significantly decreased in hNS/PCs + CM-NPs group compared to the control groups. Taken together, this study indicated that combination therapy of stem cells with CM-NPs can be an effective therapy for TBI.
Collapse
|
22
|
Microglia polarization in ischemic stroke: complex mechanisms and therapeutic interventions. Chin Med J (Engl) 2021; 134:2415-2417. [PMID: 34669634 PMCID: PMC8654435 DOI: 10.1097/cm9.0000000000001711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
23
|
Phenothiazine Inhibits Neuroinflammation and Inflammasome Activation Independent of Hypothermia After Ischemic Stroke. Mol Neurobiol 2021; 58:6136-6152. [PMID: 34455546 DOI: 10.1007/s12035-021-02542-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/17/2021] [Indexed: 01/01/2023]
Abstract
A depressive or hibernation-like effect of chlorpromazine and promethazine (C + P) on brain activity was reported to induce neuroprotection, with or without induced-hypothermia. However, the underlying mechanisms remain unclear. The current study evaluated the pharmacological function of C + P on the inhibition of neuroinflammatory response and inflammasome activation after ischemia/reperfusion. A total of 72 adult male Sprague-Dawley rats were subjected to 2 h middle cerebral artery occlusion (MCAO) followed by 6 or 24 h reperfusion. At the onset of reperfusion, rats received C + P (8 mg/kg) with temperature control. Brain cell death was detected by measuring CD68 and myeloperoxidase (MPO) levels. Inflammasome activation was measured by mRNA levels of NLRP3, IL-1β, and TXNIP, and protein quantities of NLRP3, IL-1β, TXNIP, cleaved-Caspase-1, and IL-18. Activation of JAK2/STAT3 pathway was detected by the phosphorylation of STAT3 (p-STAT3) and JAK2 (p-JAK2), and the co-localization of p-STAT3 and NLRP3. Activation of the p38 pathway was assessed with the protein levels of p-p38/p38. The mRNA and protein levels of HIF-1α, FoxO1, and p-FoxO1, and the co-localization of p-STAT3 with HIF-1α or FoxO1 were quantitated. As expected, C + P significantly reduced cell death and attenuated the neuroinflammatory response as determined by reduced CD68 and MPO. C + P decreased ischemia-induced inflammasome activation, shown by reduced mRNA and protein expressions of NLRP3, IL-1β, TXNIP, cleaved-Caspase-1, and IL-18. Phosphorylation of JAK2/STAT3 and p38 pathways and the co-localization of p-STAT3 with NLRP3 were also inhibited by C + P. Furthermore, mRNA levels of HIF-1α and FoxO1 were decreased in the C + P group. While C + P inhibited HIF-1α protein expression, it increased FoxO1 phosphorylation, which promoted the exclusion of FoxO1 from the nucleus and inhibited FoxO1 activity. At the same time, C + P reduced the co-localization of p-STAT3 with HIF-1α or FoxO1. In conclusion, C + P treatment conferred neuroprotection in stroke by suppressing neuroinflammation and NLRP3 inflammasome activation. The present study suggests that JAK2/STAT3/p38/HIF-1α/FoxO1 are vital regulators and potential targets for efficacious therapy following ischemic stroke.
Collapse
|
24
|
Rawat PS, Jaiswal A, Khurana A, Bhatti JS, Navik U. Doxorubicin-induced cardiotoxicity: An update on the molecular mechanism and novel therapeutic strategies for effective management. Biomed Pharmacother 2021; 139:111708. [PMID: 34243633 DOI: 10.1016/j.biopha.2021.111708] [Citation(s) in RCA: 427] [Impact Index Per Article: 106.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/06/2022] Open
Abstract
Doxorubicin (Dox) is a secondary metabolite of the mutated strain of Streptomyces peucetius var. Caesius and belongs to the anthracyclines family. The anti-cancer activity of Dox is mainly exerted through the DNA intercalation and inhibiting topoisomerase II enzyme in fast-proliferating tumors. However, Dox causes cumulative and dose-dependent cardiotoxicity, which results in increased risks of mortality among cancer patients and thus limiting its wide clinical applications. There are several mechanisms has been proposed for doxorubicin-induced cardiotoxicity and oxidative stress, free radical generation and apoptosis are most widely reported. Apart from this, other mechanisms are also involved in Dox-induced cardiotoxicity such as impaired mitochondrial function, a perturbation in iron regulatory protein, disruption of Ca2+ homeostasis, autophagy, the release of nitric oxide and inflammatory mediators and altered gene and protein expression that involved apoptosis. Dox also causes downregulation of DNA methyltransferase 1 (DNMT1) enzyme activity which leads to a reduction in the DNA methylation process. This hypomethylation causes dysregulation in the mitochondrial genes like peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1-alpha (PGC-1α), nuclear respiratory factor 1 (NRF-1) and mitochondrial transcription factor A (TFAM) unit in the heart. Apart from DNA methylation, Dox treatment also alters the micro RNAs levels and histone deacetylase (HDAC) activity. Therefore, in the current review, we have provided a detailed update on the current understanding of the pathological mechanisms behind the well-known Dox-induced cardiotoxicity. Further, we have provided some of the most plausible pharmacological strategies which have been tested against Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Pushkar Singh Rawat
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Aiswarya Jaiswal
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Amit Khurana
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, PVNRTVU, Rajendranagar, Hyderabad 500030, Telangana, India; Centre for Biomedical Engineering (CBME), Indian Institute of Technology (IIT), Delhi 110016, India.
| | - Jasvinder Singh Bhatti
- Department of human genetics and molecular medicine, School of health sciences, Central University of Punjab, Bathinda 151401, Punjab, India.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
25
|
Normobaric Oxygen (NBO) Therapy Reduces Cerebral Ischemia/Reperfusion Injury through Inhibition of Early Autophagy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:7041290. [PMID: 34306153 PMCID: PMC8263229 DOI: 10.1155/2021/7041290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/31/2021] [Accepted: 06/20/2021] [Indexed: 11/17/2022]
Abstract
Objectives Normobaric oxygen (NBO) therapy has great clinical potential in the treatment of ischemic stroke, but its underlying mechanism is unknown. Our study aimed to investigate the role of autophagy during the application of NBO on cerebral ischemia/reperfusion injury. Methods Male Sprague Dawley rats received 2 hours of middle cerebral artery occlusion (MCAO), followed by 2, 6, or 24 hours of reperfusion. At the beginning of reperfusion, rats were randomly given NBO (95% O2) or room air (21% O2) for 2 hours. In some animals, 3-methyladenine (3-MA, autophagy inhibitor) was administered 10 minutes before reperfusion. The severity of the ischemic injury was determined by infarct volume, neurological deficit, and apoptotic cell death. Western blotting was used to determine the protein expression of autophagy and apoptosis, while mRNA expression of apoptotic molecules was detected by real-time PCR. Results NBO treatment after ischemia/reperfusion significantly decreased infarct volume and neurobehavioral defects. The increased expression of the autophagy markers, including microtubule-associated protein 1A light chain 3 (LC3) and Beclin 1, after ischemia/reperfusion was reversed by NBO, while promoting Sequestosome 1 (p62/SQSTM1) expression. In addition, NBO reduced cerebral apoptosis in association with alleviated BAX expression and increased BCL-2 expression. 3-MA reduced autophagy and apoptotic death but did not further improve NBO-attenuated ischemic damage. Conclusion NBO induced remarkable neuroprotection from ischemic injury, which was correlated with blocked autophagy activity.
Collapse
|
26
|
Ashayeri Ahmadabad R, Mirzaasgari Z, Gorji A, Khaleghi Ghadiri M. Toll-Like Receptor Signaling Pathways: Novel Therapeutic Targets for Cerebrovascular Disorders. Int J Mol Sci 2021; 22:ijms22116153. [PMID: 34200356 PMCID: PMC8201279 DOI: 10.3390/ijms22116153] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Toll-like receptors (TLRs), a class of pattern recognition proteins, play an integral role in the modulation of systemic inflammatory responses. Cerebrovascular diseases (CVDs) are a group of pathological conditions that temporarily or permanently affect the brain tissue mostly via the decrease of oxygen and glucose supply. TLRs have a critical role in the activation of inflammatory cascades following hypoxic-ischemic events and subsequently contribute to neuroprotective or detrimental effects of CVD-induced neuroinflammation. The TLR signaling pathway and downstream cascades trigger immune responses via the production and release of various inflammatory mediators. The present review describes the modulatory role of the TLR signaling pathway in the inflammatory responses developed following various CVDs and discusses the potential benefits of the modulation of different TLRs in the improvement of functional outcomes after brain ischemia.
Collapse
Affiliation(s)
- Rezan Ashayeri Ahmadabad
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran; (R.A.A.); (Z.M.)
| | - Zahra Mirzaasgari
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran; (R.A.A.); (Z.M.)
- Department of Neurology, Iran University of Medical Sciences, Tehran 1593747811, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran; (R.A.A.); (Z.M.)
- Epilepsy Research Center, Westfälische Wilhelms-Universität, 48149 Münster, Germany
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Department of Neurosurgery, Westfälische Wilhelms-Universität, 48149 Münster, Germany;
- Department of Neurology, Westfälische Wilhelms-Universität Münster, 48149 Münster, Germany
- Correspondence: ; Tel.: +49-251-8355564; Fax: +49-251-8347479
| | | |
Collapse
|
27
|
Jiang Q, Stone CR, Elkin K, Geng X, Ding Y. Immunosuppression and Neuroinflammation in Stroke Pathobiology. Exp Neurobiol 2021; 30:101-112. [PMID: 33972464 PMCID: PMC8118752 DOI: 10.5607/en20033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Over the preceding decades, there have been substantial advances in our knowledge of the pathophysiology of stroke. One such advance has been an increased understanding of the multifarious crosstalk in which the nervous and immune systems engage in order to maintain homeostasis. By interrupting the immune-nervous nexus, it is thought that stroke induces change in both systems. Additionally, it has been found that both innate and adaptive immunosuppression play protective roles against the effects of stroke. The release of danger-/damage-associated molecular patterns (DAMPs) activates Toll-like receptors (TLRs), contributing to the harmful inflammatory effects of ischemia/reperfusion injury after stroke; the Tyro3, Axl, and MerTK (TAM)/Gas6 system, however, has been shown to suppress inflammation via downstream signaling molecules that inhibit TLR signaling. Anti-inflammatory cytokines have also been found to promote neuroprotection following stroke. Additionally, adaptive immunosuppression merits further consideration as a potential endogenous protective mechanism. In this review, we highlight recent studies regarding the effects and mechanism of immunosuppression on the pathophysiology of stroke, with the hope that a better understanding of the function of both of innate and adaptive immunity in this setting will facilitate the development of effective therapies for post-stroke inflammation.
Collapse
Affiliation(s)
- Qian Jiang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China.,Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China
| | - Christopher R Stone
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit 48201, MI, USA
| | - Kenneth Elkin
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit 48201, MI, USA
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China.,Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit 48201, MI, USA
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit 48201, MI, USA.,Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit 48201, MI, USA
| |
Collapse
|
28
|
Liu Y, Zhu X, Tong X, Tan Z. Syringin protects against cerebral ischemia/reperfusion injury via inhibiting neuroinflammation and TLR4 signaling. Perfusion 2021; 37:562-569. [PMID: 33832376 DOI: 10.1177/02676591211007025] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Cerebral ischemia/reperfusion injury (CI/R) is associated with high mortality and remains a large challenge in the clinic. Syringin is a bioactive compound with anti-inflammation, antioxidant, as well as neuroprotective effects. Nevertheless, whether syringin could protect against CI/R injury and its potential mechanism was still unclear. METHODS Rats were randomly divided into five groups: sham group, syringin group, CI/R group, CI/R + syringin group, and CI/R + syringin + LPS (TLR4 agonist) group. The CI/R injury rat model was established by the middle cerebral artery occlusion (MCAO). The learning and memory ability of rats was estimated by the Morris water maze test. Modified neurological severity score test (mNSS) and infarct volume were detected to assess the neuroprotective effect of syringin. ELISA and RT-qPCR were used to analyze the concentration of proinflammation cytokines and the expression of TLR4. RESULTS CI/R injury induced increased mNSS scores and decreased learning and memory ability of rats. Syringin could significantly protect against CI/R injury as it decreased the cerebral damage and improved the cognitive ability of CI/R rats. Moreover, syringin also reduced neuroinflammation of CI/R injury rats. Additionally, TLR4 was significantly upregulated in CI/R injury rats, which was suppressed by syringin. The activation of TLR4 reversed the neuroprotective effect of syringin in CI/R rats. CONCLUSION Syringin decreased the inflammation reaction and cerebral damage in CI/R injury rats. The neuroprotective effect of syringin may be correlated with the inhibition of TLR4.
Collapse
Affiliation(s)
- Yan Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Mudanjiang University, Mudanjiang, Heilongjiang, China
| | - Xuyao Zhu
- Department of Imaging, Hongqi Hospital affiliated to Mudanjiang Medical College, Mudanjiang, Heilongjiang, China
| | - Xiuxia Tong
- Department of Emergency, The Second Affliated Hospital of Mudanjiang University, Mudanjiang, Heilongjiang, China
| | - Ziqiang Tan
- Department of Pharmacy, The Second Affliated Hospital of Mudanjiang University, Mudanjiang, Heilongjiang, China
| |
Collapse
|
29
|
Hou K, Li G, Yu J, Xu K, Wu W. Receptors, Channel Proteins, and Enzymes Involved in Microglia-mediated Neuroinflammation and Treatments by Targeting Microglia in Ischemic Stroke. Neuroscience 2021; 460:167-180. [PMID: 33609636 DOI: 10.1016/j.neuroscience.2021.02.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 12/12/2022]
Abstract
Stroke is the largest contributor to global neurological disability-adjusted life-years, posing a huge economic and social burden to the world. Though pharmacological recanalization with recombinant tissue plasminogen activator and mechanical thrombectomy have greatly improved the prognosis of patients with ischemic stroke, clinically, there is still no effective treatment for the secondary injury caused by cerebral ischemia. In recent years, more and more evidences show that neuroinflammation plays a pivotal role in the pathogenesis and progression of ischemic cerebral injury. Microglia are brain resident innate immune cells and act the role peripheral macrophages. They play critical roles in mediating neuroinflammation after ischemic stroke. Microglia-mediated neuroinflammation is not an isolated process and has complex relationships with other pathophysiological processes as oxidative/nitrative stress, excitotoxicity, necrosis, apoptosis, pyroptosis, autophagy, and adaptive immune response. Upon activation, microglia differentially express various receptors, channel proteins, and enzymes involved in promoting or inhibiting the inflammatory processes, making them the targets of intervention for ischemic stroke. To inhibit microglia-related neuroinflammation and promote neurological recovery after ischemic stroke, numerous biochemical agents, cellular therapies, and physical methods have been demonstrated to have therapeutic potentials. Though accumulating experimental evidences have demonstrated that targeting microglia is a promising approach in the treatment of ischemic stroke, the clinical progress is slow. Till now, no clinical study could provide convincing evidence that any biochemical or physical therapies could exert neuroprotective effect by specifically targeting microglia following ischemic stroke.
Collapse
Affiliation(s)
- Kun Hou
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Guichen Li
- Department of Neurology, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Jinlu Yu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Kan Xu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Wei Wu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| |
Collapse
|
30
|
Ge F, Shao G, Chen S, Sun Y, Xu H. Chrysoeriol promotes functional neurological recovery in a rat model of cerebral ischemia. Pharmacogn Mag 2021. [DOI: 10.4103/pm.pm_329_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|