1
|
Sharma B, Purohit R. Enhanced sampling simulations to explore himalayan phytochemicals as potential phosphodiesterase-1 inhibitor for neurological disorders. Biochem Biophys Res Commun 2025; 758:151614. [PMID: 40112537 DOI: 10.1016/j.bbrc.2025.151614] [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: 12/27/2024] [Revised: 02/25/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025]
Abstract
The rising incidence of neurological and neuropsychiatric disorders underscores the urgent need for innovative and evidence based treatment strategies. Phosphodiesterase-1 (PDE1) is a dual-substrate (cAMP/cGMP) phosphodiesterase expressed in the central nervous system and peripheral areas, which modulates cyclic nucleotide signaling cascades. Inhibiting PDE1 enhances cAMP/cGMP levels, promoting neuronal plasticity and neuroprotection, making it a promising therapeutic strategy for neurological disorders. The pursuit of targeting this enzyme for treating neurological and neuropsychiatric disorders has faced obstacles due to the absence of potent, selective, and brain-penetrating inhibitors. This study aimed to identify potent PDE1 inhibitors by leveraging a diverse collection of bioactive molecules derived from Himalayan flora through computational screening methods. The four most promising hit molecules were chosen for further investigation and subjected to Molecular Dynamics (MD) simulations, binding free energy calculations, along with standard molecules. It was found that the hit molecules stigmast-7, corilagin and emblicanin-A had formed the most stable complexes, and also, the least binding free energy was observed for stigmast-7 among the hit molecules. Additionally, the pulling simulations indicated that stigmast-7 and corilagin were the most robust binders, and required the highest force to dissociate from the binding cavity completely. The umbrella sampling simulations also revealed the lowest binding free energy for corilagin and stigmast-7. The insights gained from this study provide a foundation for future research into PDE1-targeted therapies, highlighting the potential of Himalayan bioactive compounds in developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Bhanu Sharma
- Structural Bioinformatics Lab, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP, 176061, India; Academy of Scientific and Innovative Research, Ghaziabad, 201002, India
| | - Rituraj Purohit
- Structural Bioinformatics Lab, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP, 176061, India; Academy of Scientific and Innovative Research, Ghaziabad, 201002, India.
| |
Collapse
|
2
|
Pérez-Núñez R, González MF, Avalos AM, Leyton L. Impacts of PI3K/protein kinase B pathway activation in reactive astrocytes: from detrimental effects to protective functions. Neural Regen Res 2025; 20:1031-1041. [PMID: 38845231 PMCID: PMC11438337 DOI: 10.4103/nrr.nrr-d-23-01756] [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: 10/24/2023] [Revised: 04/07/2024] [Accepted: 05/06/2024] [Indexed: 07/12/2024] Open
Abstract
Astrocytes are the most abundant type of glial cell in the central nervous system. Upon injury and inflammation, astrocytes become reactive and undergo morphological and functional changes. Depending on their phenotypic classification as A1 or A2, reactive astrocytes contribute to both neurotoxic and neuroprotective responses, respectively. However, this binary classification does not fully capture the diversity of astrocyte responses observed across different diseases and injuries. Transcriptomic analysis has revealed that reactive astrocytes have a complex landscape of gene expression profiles, which emphasizes the heterogeneous nature of their reactivity. Astrocytes actively participate in regulating central nervous system inflammation by interacting with microglia and other cell types, releasing cytokines, and influencing the immune response. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway is a central player in astrocyte reactivity and impacts various aspects of astrocyte behavior, as evidenced by in silico , in vitro , and in vivo results. In astrocytes, inflammatory cues trigger a cascade of molecular events, where nuclear factor-κB serves as a central mediator of the pro-inflammatory responses. Here, we review the heterogeneity of reactive astrocytes and the molecular mechanisms underlying their activation. We highlight the involvement of various signaling pathways that regulate astrocyte reactivity, including the PI3K/AKT/mammalian target of rapamycin (mTOR), α v β 3 integrin/PI3K/AKT/connexin 43, and Notch/PI3K/AKT pathways. While targeting the inactivation of the PI3K/AKT cellular signaling pathway to control reactive astrocytes and prevent central nervous system damage, evidence suggests that activating this pathway could also yield beneficial outcomes. This dual function of the PI3K/AKT pathway underscores its complexity in astrocyte reactivity and brain function modulation. The review emphasizes the importance of employing astrocyte-exclusive models to understand their functions accurately and these models are essential for clarifying astrocyte behavior. The findings should then be validated using in vivo models to ensure real-life relevance. The review also highlights the significance of PI3K/AKT pathway modulation in preventing central nervous system damage, although further studies are required to fully comprehend its role due to varying factors such as different cell types, astrocyte responses to inflammation, and disease contexts. Specific strategies are clearly necessary to address these variables effectively.
Collapse
Affiliation(s)
- Ramón Pérez-Núñez
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - María Fernanda González
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
3
|
Wang Y, Li M, Jiang Y, Ji Q. Comparative efficacy of neuroprotective agents for improving neurological function and prognosis in acute ischemic stroke: a network meta-analysis. Front Neurosci 2025; 18:1530987. [PMID: 39834702 PMCID: PMC11743486 DOI: 10.3389/fnins.2024.1530987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Background Ischemic stroke is the second leading cause of death and the third leading cause of combined disability and mortality globally. While reperfusion therapies play a critical role in the management of acute ischemic stroke (AIS), their applicability is limited, leaving many patients with significant neurological deficits and poor prognoses. Neuroprotective agents have garnered attention for their potential as adjunct therapies; however, their relative efficacy remains unclear. This study utilized a network meta-analysis (NMA) to systematically compare the efficacy of neuroprotective agents in improving neurological function and prognosis in stroke patients. Methods This study adhered to PRISMA guidelines and the Cochrane Handbook for systematic reviews. Randomized controlled trials (RCTs) were identified through comprehensive searches of the PubMed, Embase, and Cochrane Library databases. Two independent reviewers conducted the selection process, data extraction, and quality assessment. Outcomes included 90-day modified Rankin Scale (90d-mRS), change of National Institutes of Health Stroke Scale score from baseline to 90-day/14-day/7-day (90d/14d/7d-NIHSS) and 90-day/14-day Barthel Index (90d/14d-BI). Data analyses were performed using RevMan 5.4 and Stata 14.0. Results A total of 42 RCTs involving 12,210 participants were included in this analysis. The interventions assessed included Cerebrolysin, Citicoline, Edaravone, Edaravone Dextranol, Human urinary kallidinogenase, Minocycline, Nerinetide, Butylphthalide, Vinpocetine, and Control. The NMA results demonstrated that NBP ranked highest for the 90d-mRS, 90d-NIHSS, 14d-NIHSS, and 14d-BI outcomes. Edaravone was found to be the most effective intervention for the 7d-NIHSS and 90d-BI outcomes. Conclusion The findings of this study indicate that different neuroprotective agents exhibit distinct advantages at specific stages of recovery. NBP showed outstanding performance in improving 90d-mRS and 90d-NIHSS, underscoring its potential in long-term rehabilitation. Edaravone demonstrated significant superiority in 7d-NIHSS scores, highlighting its role in early neuroprotection. These results provide valuable insights for individualized clinical treatment. To further validate the efficacy and safety of neuroprotective agents, future studies should involve larger sample sizes and conduct multicenter, large-scale randomized controlled trials. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=601346, identifier CRD42024601346.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Mengqi Li
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Yuye Jiang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Qiuhong Ji
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
4
|
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
|
5
|
Rong J, Yamasaki T, Chen J, Kumata K, Zhao C, Fujinaga M, Hu K, Mori W, Zhang Y, Xie L, Chaudhary AF, Zhou X, Zhang W, Gao Y, Zhang K, Patel JS, Song Z, Collier TL, Yuan H, Ran C, Haider A, Li Y, Zhang MR, Liang S. Development of a Candidate 11C-Labeled Selective Phosphodiesterase 1 Radioligand for Positron Emission Tomography. ACS OMEGA 2024; 9:44154-44163. [PMID: 39524622 PMCID: PMC11541501 DOI: 10.1021/acsomega.4c03214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Phosphodiesterases (PDEs) constitute a superfamily of phosphohydrolytic enzymes that regulate intracellular second messenger signaling by hydrolyzing cyclic adenosine monophosphate and cyclic guanosine monophosphate. Among the 11 subfamilies of PDEs, phosphodiesterase 1 (PDE1) stands out due to its broad implications in central and peripheral pathologies. There are three subtypes of PDE1: PDE1A, PDE1B, and PDE1C. While PDE1A and PDE1C are distributed in both the brain and peripheral organs, PDE1B is predominantly expressed in the brain, rendering it an attractive drug target for neurological and psychological disorders. Despite continuous efforts dedicated to the development of novel PDE1 inhibitors, a suitable PDE1 radioligand for human use is currently lacking. In this study, we present the identification and preclinical evaluation of [11C]PF-04822163, a selective radioligand candidate for imaging PDE1 with positron emission tomography. PF-04822163 exhibits excellent potency toward PDE1 and demonstrates great target selectivity over other PDEs. Then, PF-04822163 was labeled with carbon-11 (half-life, 20 min) in favorable radiochemical yields (25 ± 10%, decay-corrected) and high molar activities (106-194 GBq/μmol). Further, in vitro and in vivo evaluations in rodents suggested that [11C]PF-04822163 displayed good brain penetration and a rapid washout. Despite these promising performance characteristics of [11C]PF-04822163, only marginal specific binding was observed in vivo. Further optimization of the scaffold is warranted to obtain favorable pharmacological and ADME properties.
Collapse
Affiliation(s)
- Jian Rong
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital,
and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Tomoteru Yamasaki
- Department
of Advanced Nuclear Medicine Sciences, Institute
for Quantum Medical Science, National Institutes for Quantum Science
and Technology, Chiba 263-8555, Japan
| | - Jiahui Chen
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital,
and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Katsushi Kumata
- Department
of Advanced Nuclear Medicine Sciences, Institute
for Quantum Medical Science, National Institutes for Quantum Science
and Technology, Chiba 263-8555, Japan
| | - Chunyu Zhao
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital,
and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Masayuki Fujinaga
- Department
of Advanced Nuclear Medicine Sciences, Institute
for Quantum Medical Science, National Institutes for Quantum Science
and Technology, Chiba 263-8555, Japan
| | - Kuan Hu
- Department
of Advanced Nuclear Medicine Sciences, Institute
for Quantum Medical Science, National Institutes for Quantum Science
and Technology, Chiba 263-8555, Japan
| | - Wakana Mori
- Department
of Advanced Nuclear Medicine Sciences, Institute
for Quantum Medical Science, National Institutes for Quantum Science
and Technology, Chiba 263-8555, Japan
| | - Yiding Zhang
- Department
of Advanced Nuclear Medicine Sciences, Institute
for Quantum Medical Science, National Institutes for Quantum Science
and Technology, Chiba 263-8555, Japan
| | - Lin Xie
- Department
of Advanced Nuclear Medicine Sciences, Institute
for Quantum Medical Science, National Institutes for Quantum Science
and Technology, Chiba 263-8555, Japan
| | - Ahmad F. Chaudhary
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
| | - Xin Zhou
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
| | - Wei Zhang
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
| | - Yabiao Gao
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
| | - Kuo Zhang
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
| | - Jimmy S. Patel
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Department
of Radiation Oncology, Winship Cancer Institute
of Emory University, Atlanta, Georgia 30322, United States
| | - Zhendong Song
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
| | - Thomas L. Collier
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital,
and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Hongjie Yuan
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, Atlanta, Georgia 30322, United States
| | - Chongzhao Ran
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Ahmed Haider
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital,
and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Yinlong Li
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital,
and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Ming-Rong Zhang
- Department
of Advanced Nuclear Medicine Sciences, Institute
for Quantum Medical Science, National Institutes for Quantum Science
and Technology, Chiba 263-8555, Japan
| | - Steven Liang
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital,
and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
| |
Collapse
|
6
|
Kumari S, Dhapola R, Sharma P, Nagar P, Medhi B, HariKrishnaReddy D. The impact of cytokines in neuroinflammation-mediated stroke. Cytokine Growth Factor Rev 2024; 78:105-119. [PMID: 39004599 DOI: 10.1016/j.cytogfr.2024.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024]
Abstract
Cerebral stroke is ranked as the third most common contributor to global mortality and disability. The involvement of inflammatory mechanisms, both peripherally and within the CNS, holds significance in the pathophysiological cascades following the initiation of stroke. After the onset of acute stroke, predominantly ischemic, a subsequent phase of neuroinflammation ensues. It is a dual-effect process that not only exacerbates injury, leading to cell death, but paradoxically, it also serves a shielding role in facilitating recovery. Cytokines serve as pivotal mediators within the inflammatory cascade, actively contributing to the progression of ischemic damage. Stroke is followed by increased expression of pro-inflammatory cytokines including TNF-α, IL-1β, IL-6, etc. leading to the recruitment and stimulation of glial cells and peripheral leukocytes at the site of injury, promoting neuroinflammation. Cytokines can directly induce neuronal injury and death through various mechanisms, including excitotoxicity, oxidative stress, HPA-axis activation, secretion of matrix metalloproteinase and apoptosis. They can also amplify the inflammatory response, leading to further neuronal damage. Therapeutic strategies aimed at modulating cytokine release, immune response and cytokine signalling or activity are being explored as potential interventions to mitigate neuroinflammation and its detrimental effects in stroke. In this review, we have given a concise summary of our current knowledge of the function of various cytokines, brain inflammation and various signalling and molecular pathways including JAK/STAT3, TGF-β/Smad, MAPK, HMGB1/TLR and NF-κB modulated cytokines regulation in stroke. Therapeutic agents such as MCC950, genistein, edaravone, minocycline, etc. targeting various cytokines-associated signalling pathways have shown efficacy in preclinical and clinical trials reducing the pathophysiology of the illness were also addressed in this study.
Collapse
Affiliation(s)
- Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Pushank Nagar
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India.
| |
Collapse
|
7
|
Dong ZC, Shi Y, Liu LJ, Feng TT, Zhou Y, Pan BW. Synthesis and pharmacological activity of vinpocetine derivatives. RSC Adv 2024; 14:7981-7991. [PMID: 38454939 PMCID: PMC10918451 DOI: 10.1039/d3ra07325d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Vinpocetine and its derivatives were extensively employed in the treatment of ischemic stroke, serving as effective cerebrovascular vasodilators. They could also be utilized for neuroprotection, anti-inflammatory purposes, anti-aging interventions, insomnia treatment, and antidepressant effects. However, due to issues such as hepatic first-pass effect, low bioavailability, and poor patient compliance with multiple dosing, the secondary development of Vinpocetine to address these limitations became a prominent area of research. Five primary methodologies were employed for the synthesis of Vinpocetine derivatives. These included substitution on the A ring to modify the 14-ester group, alteration of the 16-ethyl group, simplification of the D and E rings, and modification of the conformation of Vinpocetine. This paper summarized the current synthesis and activity studies of Vinpocetine and its derivatives, with the aim of providing a reference for the discovery of more potent derivatives of Vinpocetine.
Collapse
Affiliation(s)
- Zhang Chao Dong
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine Guiyang 550025 China
| | - Yang Shi
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine Guiyang 550025 China
| | - Li Juan Liu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine Guiyang 550025 China
| | - Ting Ting Feng
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine Guiyang 550025 China
| | - Ying Zhou
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine Guiyang 550025 China
| | - Bo Wen Pan
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine Guiyang 550025 China
| |
Collapse
|
8
|
Xu G, Dong F, Su L, Tan ZX, Lei M, Li L, Wen D, Zhang F. The role and therapeutic potential of nuclear factor κB (NF-κB) in ischemic stroke. Biomed Pharmacother 2024; 171:116140. [PMID: 38211425 DOI: 10.1016/j.biopha.2024.116140] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Stroke is a prevalent cerebrovascular condition with a global impact, causing significant rates of illness and death. Despite extensive research, the available treatment options for stroke remain restricted. Hence, it is crucial to gain a deeper understanding of the molecular mechanisms associated with the onset and advancement of stroke in order to establish a theoretical foundation for novel preventive and therapeutic approaches. NF-κB, also known as nuclear factor κB, is a transcription factor responsible for controlling the expression of numerous genes and plays a crucial role in diverse physiological processes. NF-κB is triggered and regulates neuroinflammation and other processes after stroke, promoting the generation of cytokine storms and contributing to the advancement of ischemic stroke (IS). Therefore, NF-κB could potentially play a vital role in stroke by regulating diverse pathophysiological processes. This review provides an overview of the functions of NF-κB in stroke and its governing mechanisms. In addition, our attention is directed towards various potential therapies that aim to inhibit the NF-κB signaling pathway in order to offer valuable insights for the advancement of innovative treatment approaches for stroke.
Collapse
Affiliation(s)
- Guangyu Xu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lei Su
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding 071000, PR China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Mingcheng Lei
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lina Li
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, PR China; Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, PR China.
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China.
| |
Collapse
|
9
|
Salman HR, Alzubaidy AA, Abbas AH, Mohammad HA. Attenuated effects of topical vinpocetine in an imiquimod-induced mouse model of psoriasis. J Taibah Univ Med Sci 2024; 19:35-53. [PMID: 37868105 PMCID: PMC10585306 DOI: 10.1016/j.jtumed.2023.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 08/11/2023] [Accepted: 09/09/2023] [Indexed: 10/24/2023] Open
Abstract
Psoriasis is an uncontrolled, long-lasting inflammatory dermatosis distinguished by thickened, erythematous, and flaky skin lesions. Massive amounts of inflammatory cytokines are produced when immune system imbalances are driven by genetic and environmental triggers. Vinpocetine (VNP), a man-made analogue of the compound vincamine found in the dwarf periwinkle herb, has robust anti-inflammatory, immunomodulatory, and anti-oxidative effects; alleviates the epidermal penetration of immune cells, such as eosinophils and neutrophils; and abolishes the generation of pro-inflammatory molecules. Objective This study was aimed at exploring the effects of long-term topical VNP, both alone and co-administered with clobetasol propionate, in an imiquimod-induced mouse model of psoriasiform dermatitis. Methods The study protocol consisted of 48 Swiss albino mice, randomly divided into six groups of eight mice each. In group I, petroleum jelly was administered daily for 8 days. In group II, imiquimod was administered topically at 62.5 mg daily for 8 days. In groups III, VI, V, and VI, 0.05% clobetasol propionate, 1% VNP, 3% VNP, and 3% VNP plus 0.05% clobetasol were administered topically for an additional 8 days after the induction, thus resulting in a total trial length of 16 days. Results Topical VNP at various doses alleviated the severity of imiquimod-induced psoriatic lesions-including erythema, silvery-white scaling, and thickening-and reversed the histopathological abnormalities. Moreover, imiquimod-exposed animals treated with VNP showed markedly diminished concentrations of inflammatory biomarkers, including tumour necrosis factor-α, interleukin (IL)-8, IL-17A, IL-23, IL-37, nuclear factor-kappa B (NF-κB), and transforming growth factor-β1. Conclusion This research provides new evidence that VNP, alone and in combination with clobetasol, may serve as a potential adjuvant for long-term management of autoimmune and autoinflammatory skin diseases, particularly psoriasis, by attenuating psoriatic lesion severity, suppressing cytokine generation, and limiting NF-κB-mediated inflammation.
Collapse
Affiliation(s)
- Hayder R. Salman
- Al-Mustaqbal University, College of Pharmacy, Department of Pharmacology, Hillah, Babylon, Iraq
- Al-Nahrain University, College of Medicine, Department of Pharmacology, Baghdad, Iraq
| | - Adeeb A. Alzubaidy
- University of Warith Al-Anbiyaa, College of Medicine, Department of Pharmacology, Karbala, Iraq
| | - Alaa H. Abbas
- Al-Nahrain University, College of Medicine, Department of Pharmacology, Baghdad, Iraq
| | - Hussein A. Mohammad
- University of Al-Qadisiyah, College of Pharmacy, Department of Pharmaceutics, Al Diwaniya, Al-Qadisiyah Province, Iraq
| |
Collapse
|
10
|
Zhang W, Xu H, Li C, Han B, Zhang Y. Exploring Chinese herbal medicine for ischemic stroke: insights into microglia and signaling pathways. Front Pharmacol 2024; 15:1333006. [PMID: 38318134 PMCID: PMC10838993 DOI: 10.3389/fphar.2024.1333006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024] Open
Abstract
Ischemic stroke is a prevalent clinical condition affecting the central nervous system, characterized by a high mortality and disability rate. Its incidence is progressively rising, particularly among younger individuals, posing a significant threat to human well-being. The activation and polarization of microglia, leading to pro-inflammatory and anti-inflammatory responses, are widely recognized as pivotal factors in the pathogenesis of cerebral ischemia and reperfusion injury. Traditional Chinese herbal medicines (TCHMs) boasts a rich historical background, notable efficacy, and minimal adverse effects. It exerts its effects by modulating microglia activation and polarization, suppressing inflammatory responses, and ameliorating nerve injury through the mediation of microglia and various associated pathways (such as NF-κB signaling pathway, Toll-like signaling pathway, Notch signaling pathway, AMPK signaling pathway, MAPK signaling pathway, among others). Consequently, this article focuses on microglia as a therapeutic target, reviewing relevant pathway of literature on TCHMs to mitigate neuroinflammation and mediate IS injury, while also exploring research on drug delivery of TCHMs. The ultimate goal is to provide new insights that can contribute to the clinical management of IS using TCHMs.
Collapse
Affiliation(s)
| | | | | | - Bingbing Han
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yimin Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
11
|
Zhu Z, Tang W, Qiu X, Xin X, Zhang J. Advances in targeting Phosphodiesterase 1: From mechanisms to potential therapeutics. Eur J Med Chem 2024; 263:115967. [PMID: 38000211 DOI: 10.1016/j.ejmech.2023.115967] [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: 10/04/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023]
Abstract
Phosphodiesterase 1 (PDE1) is an enzyme entrusted with the hydrolysis of the second messengers cAMP and cGMP, thereby governing a plethora of metabolic processes, encompassing ion channel modulation and cellular apoptosis. Recent advancements in the realm of small molecule structural variations have greatly facilitated the exploration of innovative applications for PDE1. Remarkably, a recent series of PDE1 inhibitors (PDE1i) have been meticulously formulated and devised, showcasing enhanced selectivity and potency. Among them, ITI-214 has entered Phase II clinical trials, holding promise for the treatment of Parkinson's disease and heart failure. Nevertheless, the majority of current PDE1 inhibitors have encountered substantial side effects in clinical trials attributable to their limited selectivity, this predicament presents a formidable obstacle in the development of specific small molecule inhibitors targeting PDE1. This Perspective endeavors to illuminate the potential design approaches, structure-activity relationships, and biological activities of current PDE1i, aiming to offer support and insights for clinical practice and the development of novel PDE1i.
Collapse
Affiliation(s)
- Ziyu Zhu
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wentao Tang
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xuemei Qiu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xin Xin
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
12
|
Lin W, Zhao XY, Cheng JW, Li LT, Jiang Q, Zhang YX, Han F. Signaling pathways in brain ischemia: Mechanisms and therapeutic implications. Pharmacol Ther 2023; 251:108541. [PMID: 37783348 DOI: 10.1016/j.pharmthera.2023.108541] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Ischemic stroke occurs when the arteries supplying blood to the brain are narrowed or blocked, inducing damage to brain tissue due to a lack of blood supply. One effective way to reduce brain damage and alleviate symptoms is to reopen blocked blood vessels in a timely manner and reduce neuronal damage. To achieve this, researchers have focused on identifying key cellular signaling pathways that can be targeted with drugs. These pathways include oxidative/nitrosative stress, excitatory amino acids and their receptors, inflammatory signaling molecules, metabolic pathways, ion channels, and other molecular events involved in stroke pathology. However, evidence suggests that solely focusing on protecting neurons may not yield satisfactory clinical results. Instead, researchers should consider the multifactorial and complex mechanisms underlying stroke pathology, including the interactions between different components of the neurovascular unit. Such an approach is more representative of the actual pathological process observed in clinical settings. This review summarizes recent research on the multiple molecular mechanisms and drug targets in ischemic stroke, as well as recent advances in novel therapeutic strategies. Finally, we discuss the challenges and future prospects of new strategies based on the biological characteristics of stroke.
Collapse
Affiliation(s)
- Wen Lin
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiang-Yu Zhao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jia-Wen Cheng
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Li-Tao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Quan Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yi-Xuan Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China.
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China; Institute of Brain Science, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
13
|
Zhang Q, Jiang Q, Sa K, Liang J, Sun D, Li H, Chen L. Research progress of plant-derived natural alkaloids in central nervous system diseases. Phytother Res 2023; 37:4885-4907. [PMID: 37455555 DOI: 10.1002/ptr.7955] [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: 10/24/2022] [Revised: 05/14/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Central nervous system (CNS) disease is one of the most important causes of human death. Because of their complex pathogenesis, more and more attention has been paid to them. At present, drug treatment of the CNS is the main means; however, most drugs only relieve symptoms, and some have certain toxicity and side effects. Natural compounds derived from plants can provide safer and more effective alternatives. Alkaloids are common nitrogenous basic organic compounds found in nature, which exist widely in many kinds of plants and have unique application value in modern medicine. For example, Galantamine and Huperzine A from medicinal plants are widely used drugs on the market to treat Alzheimer's disease. Therefore, the main purpose of this review is to provide the available information on natural alkaloids with the activity of treating central nervous system diseases in order to explore the trends and perspectives for the further study of central nervous system drugs. In this paper, 120 alkaloids with the potential effect of treating central nervous system diseases are summarized from the aspects of sources, structure types, mechanism of action and structure-activity relationship.
Collapse
Affiliation(s)
- Qingqing Zhang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Qinghua Jiang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kuiru Sa
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Junming Liang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Dejuan Sun
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
14
|
Petric Z, Paixão P, Filipe A, Guimarães Morais J. Clinical Pharmacology of Vinpocetine: Properties Revisited and Introduction of a Population Pharmacokinetic Model for Its Metabolite, Apovincaminic Acid (AVA). Pharmaceutics 2023; 15:2502. [PMID: 37896263 PMCID: PMC10610279 DOI: 10.3390/pharmaceutics15102502] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/07/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
This paper examines the use of vinpocetine in the context of clinical pharmacology. The main and active metabolite of vinpocetine is apovincaminic acid (AVA). Due to the scarce information in the literature on AVA pharmacokinetics, we propose a population pharmacokinetic (PopPK) model for AVA based on a study in healthy volunteers with three different formulations of vinpocetine. The suggested PopPK model (and simulations) could be helpful in ensuring the more effective and safer use of the vinpocetine in the future given the increasing range of suggested indications for its use.
Collapse
Affiliation(s)
- Zvonimir Petric
- Department of Pharmacological Sciences, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-004 Lisboa, Portugal
| | - Paulo Paixão
- Department of Pharmacological Sciences, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-004 Lisboa, Portugal
| | - Augusto Filipe
- Medical Department, Tecnimede, Sociedade Técnico-Medicinal, S.A., Zona Industrial da Abrunheira, Rua da Tapada Grande, No. 2 Abrunheira, 2710-089 Sintra, Portugal
| | - José Guimarães Morais
- Department of Pharmacological Sciences, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-004 Lisboa, Portugal
| |
Collapse
|
15
|
Hou B, Li D, Wang D, Jiang C, Wang G, Wang D, Yan G, Guo X, Zhao L, Wan Z, Fan C, Cao W, Ren H. Neuroprotective Effects of Vinpocetine Against Ischemia-Reperfusion Injury Via Inhibiting NLRP3 Inflammasome Signaling Pathway. Neuroscience 2023; 526:74-84. [PMID: 37290685 DOI: 10.1016/j.neuroscience.2023.05.021] [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: 02/18/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/10/2023]
Abstract
Ischemic stroke is one of the main causes of serious disability and death worldwide. NLRP3 inflammasome is an intracellular pattern recognition receptor composed of polyprotein complex, which participates in mediating a series of inflammatory responses and is considered as a potential target for the treatment of ischemic stroke. Vinpocetine, a derivative of vincamine, has been widely used in the prevention and treatment of ischemic stroke. However, the therapeutic mechanism of vinpocetine is not clear, and its effect on NLRP3 inflammasome remains to be determined. In this study, we used the mouse model of transient middle cerebral artery occlusion (tMCAO) to simulate the occurrence of ischemic stroke. Different doses of vinpocetine (5, 10, 15 mg/kg/d) were injected intraperitoneally for 3 days after ischemia-reperfusion in mice. The effects of different doses of vinpocetine on the degree of ischemia-reperfusion injury in mice were observed by TTC staining and modified neurological severity score scale, and the optimal dose was determined. Then, based on this optimal dose, we observed the effects of vinpocetine on apoptosis, microglial proliferation and NLRP3 inflammasome. In addition, we compared the effects of vinpocetine and MCC950 (a specific inhibitor of NLRP3 inflammasome) on NLRP3 inflammasome. Our results show that vinpocetine can effectively reduce the infarct volume and promote the recovery of behavioral function in stroke mice, and the maximal beneficial effects were observed at the dose of 10 mg/kg/d. Vinpocetine can effectively inhibit the apoptosis of peri-infarct neurons, promote the expression of Bcl-2, inhibit the expression of Bax and Cleaved Caspase-3, and reduce the proliferation of peri-infarct microglia. In addition, vinpocetine, like MCC950, can reduce the expression of NLRP3 inflammasome. Therefore, vinpocetine can effectively alleviate the ischemia-reperfusion injury in mice, and the inhibition of NLRP3 inflammasome may be an important therapeutic mechanism of vinpocetine.
Collapse
Affiliation(s)
- Boru Hou
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.
| | - Donghai Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Dengfeng Wang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Cheng Jiang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Gang Wang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Dong Wang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Guizhong Yan
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiumei Guo
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Lixia Zhao
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhuangzhuang Wan
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Chenlong Fan
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wencheng Cao
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Haijun Ren
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
16
|
Mittal P, Dhankhar S, Chauhan S, Garg N, Bhattacharya T, Ali M, Chaudhary AA, Rudayni HA, Al-Zharani M, Ahmad W, Khan SUD, Singh TG, Mujwar S. A Review on Natural Antioxidants for Their Role in the Treatment of Parkinson's Disease. Pharmaceuticals (Basel) 2023; 16:908. [PMID: 37513820 PMCID: PMC10385773 DOI: 10.3390/ph16070908] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 07/30/2023] Open
Abstract
The neurodegenerative condition known as Parkinson's disease (PD) is brought on by the depletion of dopaminergic neurons in the basal ganglia, which is the brain region that controls body movement. PD occurs due to many factors, from which one of the acknowledged effects of oxidative stress is pathogenic pathways that play a role in the development of Parkinson's disease. Antioxidants, including flavonoids, vitamins E and C, and polyphenolic substances, help to reduce the oxidative stress brought on by free radicals. Consequently, this lowers the risk of neurodegenerative disorders in the long term. Although there is currently no cure for neurodegenerative illnesses, these conditions can be controlled. The treatment of this disease lessens its symptoms, which helps to preserve the patient's quality of life. Therefore, the use of naturally occurring antioxidants, such as polyphenols, which may be obtained through food or nutritional supplements and have a variety of positive effects, has emerged as an appealing alternative management strategy. This article will examine the extent of knowledge about antioxidants in the treatment of neurodegenerative illnesses, as well as future directions for research. Additionally, an evaluation of the value of antioxidants as neuroprotective agents will be provided.
Collapse
Affiliation(s)
- Pooja Mittal
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (S.D.); (S.C.); (T.G.S.); (S.M.)
| | - Sanchit Dhankhar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (S.D.); (S.C.); (T.G.S.); (S.M.)
- Ganpati Institute of Pharmacy, Bilaspur 135102, India
| | - Samrat Chauhan
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (S.D.); (S.C.); (T.G.S.); (S.M.)
| | - Nitika Garg
- Ganpati Institute of Pharmacy, Bilaspur 135102, India
| | - Tanima Bhattacharya
- Department of Food and Nutrition, BioNanocomposite Research Center, Kyung Hee University, 260 Kyunghee-daero, Seoul 02447, Republic of Korea
- Nondestructive Bio-Sensing Laboratory, Department of Biosystems Machinery Engineering, College of Agriculture and Life Science, Chungnam National University, 99 Daehak-ro, BLDG# E10-2, RM# 2213, Daejeon 34134, Republic of Korea
| | - Maksood Ali
- Department of Pharmacognosy, Orlean College of Pharmacy, Dr. A.P.J. Abdul Kalam Technical University, 42, Knowledge Park—III, Greater Noida 201308, India;
- Department of Pharmacognosy, HIMT College of Pharmacy, Dr. A.P.J. Abdul Kalam Technical University, 8, Institutional Area, Knowledge Park—I, Greater Noida 201301, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia; (H.A.R.); (M.A.-Z.)
| | - Hassan Ahmad Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia; (H.A.R.); (M.A.-Z.)
| | - Mohammed Al-Zharani
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia; (H.A.R.); (M.A.-Z.)
| | - Wasim Ahmad
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Dammam 34222, Saudi Arabia;
| | - Salah Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (S.D.); (S.C.); (T.G.S.); (S.M.)
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (S.D.); (S.C.); (T.G.S.); (S.M.)
| |
Collapse
|
17
|
Szepanowski RD, Haupeltshofer S, Vonhof SE, Frank B, Kleinschnitz C, Casas AI. Thromboinflammatory challenges in stroke pathophysiology. Semin Immunopathol 2023:10.1007/s00281-023-00994-4. [PMID: 37273022 DOI: 10.1007/s00281-023-00994-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/27/2023] [Indexed: 06/06/2023]
Abstract
Despite years of encouraging translational research, ischemic stroke still remains as one of the highest unmet medical needs nowadays, causing a tremendous burden to health care systems worldwide. Following an ischemic insult, a complex signaling pathway emerges leading to highly interconnected thrombotic as well as neuroinflammatory signatures, the so-called thromboinflammatory cascade. Here, we thoroughly review the cell-specific and time-dependent role of different immune cell types, i.e., neutrophils, macrophages, T and B cells, as key thromboinflammatory mediators modulating the neuroinflammatory response upon stroke. Similarly, the relevance of platelets and their tight crosstalk with a variety of immune cells highlights the relevance of this cell-cell interaction during microvascular dysfunction, neovascularization, and cellular adhesion. Ultimately, we provide an up-to-date overview of therapeutic approaches mechanistically targeting thromboinflammation currently under clinical translation, especially focusing on phase I to III clinical trials.
Collapse
Affiliation(s)
- R D Szepanowski
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
| | - S Haupeltshofer
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
| | - S E Vonhof
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
| | - B Frank
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
| | - C Kleinschnitz
- Department of Neurology, University Hospital Essen, Essen, Germany.
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany.
| | - A I Casas
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
- Department of Pharmacology and Personalised Medicine, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
18
|
Shekarian M, Salehi I, Raoufi S, Asadbegi M, Kourosh-Arami M, Komaki A. Neuroprotective effects of vinpocetine, as a phosphodiesterase 1 inhibitor, on long-term potentiation in a rat model of Alzheimer's disease. BMC Neurosci 2023; 24:20. [PMID: 36927298 PMCID: PMC10018848 DOI: 10.1186/s12868-023-00790-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Vinpocetine (Vin) is known as a phosphodiesterase 1 inhibitor (PDE1-I) drug with multilateral effects, including antioxidant and anti-inflammatory activity. In this research, we investigated the neuroprotective and therapeutic effects of Vin through hippocampal synaptic plasticity on a rat's model of Alzheimer's disease (AD) induced by an intracerebroventricular (ICV) injection of beta-amyloid (Aβ). METHODS Sixty adult male Wistar rats were randomly divided into six groups: 1. control, 2. sham, 3. Aβ, 4. pretreatment (Vin + Aβ): Vin (4 mg/kg, gavage) for 30 days and then, inducing an AD model by an ICV injection of Aβ(1-42), 5. treatment (Aβ + Vin): inducing an AD model and then receiving Vin for 30 days by gavage, and 7. pretreatment + treatment (Vin + Aβ + Vin): receiving Vin by gavage for 30 days before and 30 days after the induction of an AD model. After these procedures, via stereotaxic surgery, the stimulating electrodes were placed at the perforant pathway (PP) and the recording electrodes were implanted in the dentate gyrus. RESULTS Excitatory postsynaptic potential (EPSP) slope and population spike (PS) amplitude in the Aβ group meaningfully diminished compared to the control group after the induction of long-term potentiation (LTP). CONCLUSIONS Vin could significantly prevent the Aβ effects on LTP. It can be concluded that pretreatment and treatment with Vin can be neuroprotective against harmful consequences of Aβ on hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Meysam Shekarian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Iraj Salehi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Safoura Raoufi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Masoumeh Asadbegi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Komaki
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran.
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
19
|
Li M, Wang S, Zhang C, Chi C, Liu R, Wang T, Fu F. Escin alleviates stress-induced intestinal dysfunction to protect brain injury by regulating the gut-brain axis in ischemic stroke rats. Int Immunopharmacol 2023; 115:109659. [PMID: 36608442 DOI: 10.1016/j.intimp.2022.109659] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/15/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023]
Abstract
Hyperactivity of HPA axis results in intestinal dysfunction, which may play a role in brain injury caused by ischemic stroke (IS). Escin shows a neuroprotective effect but it may not penetrate blood brain barrier (BBB). Previous work in our laboratory showed that escin ameliorated intestinal injury in animals. The aim of this study is to investigate whether escin attenuates brain injury by improving intestinal dysfunction in middle cerebral artery occlusion (MCAO) rats, to mimic IS. MCAO rats and lipopolysaccharides (LPS)-induced Caco-2 cells were used to evaluate the effects of escin in vivo and in vitro. The results showed that escin could not penetrate BBB but reduced brain infarct volume, improved neurological function, inhibited neuroinflammation, ameliorated intestinal dysfunction and tissue integrity by increasing the expression of the tight junction protein in vivo and in vitro. Escin reduced the increased corticosterone and endotoxin level in blood of MCAO rats, regulated GR/p38 MAPK/NF-κB signaling pathway in ileal tissue and LPS/TLR4/NF-κB signaling pathway in ischemic brain tissue. These findings suggest that escin could attenuate ischemic brain injury by improving intestinal dysfunction, and it may be a promising way to protect brain injury by protecting intestine, instead of targeting the brain directly after IS.
Collapse
Affiliation(s)
- Min Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China; College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China
| | - Shengguang Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China
| | - Ce Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Chenglin Chi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Rongxia Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China.
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China.
| |
Collapse
|
20
|
Tabolacci C, De Vita D, Facchiano A, Bozzuto G, Beninati S, Failla CM, Di Martile M, Lintas C, Mischiati C, Stringaro A, Del Bufalo D, Facchiano F. Phytochemicals as Immunomodulatory Agents in Melanoma. Int J Mol Sci 2023; 24:2657. [PMID: 36768978 PMCID: PMC9916941 DOI: 10.3390/ijms24032657] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
Cutaneous melanoma is an immunogenic highly heterogenic tumor characterized by poor outcomes when it is diagnosed late. Therefore, immunotherapy in combination with other anti-proliferative approaches is among the most effective weapons to control its growth and metastatic dissemination. Recently, a large amount of published reports indicate the interest of researchers and clinicians about plant secondary metabolites as potentially useful therapeutic tools due to their lower presence of side effects coupled with their high potency and efficacy. Published evidence was reported in most cases through in vitro studies but also, with a growing body of evidence, through in vivo investigations. Our aim was, therefore, to review the published studies focused on the most interesting phytochemicals whose immunomodulatory activities and/or mechanisms of actions were demonstrated and applied to melanoma models.
Collapse
Affiliation(s)
- Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Daniela De Vita
- Department of Environmental Biology, University of Rome La Sapienza, 00185 Rome, Italy
| | | | - Giuseppina Bozzuto
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Simone Beninati
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | | | - Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico, 00128 Rome, Italy
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Carlo Mischiati
- Department of Neuroscience and Rehabilitation, School of Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Annarita Stringaro
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
21
|
Regulation of Microglia-Activation-Mediated Neuroinflammation to Ameliorate Ischemia-Reperfusion Injury via the STAT5-NF-κB Pathway in Ischemic Stroke. Brain Sci 2022; 12:brainsci12091153. [PMID: 36138889 PMCID: PMC9496994 DOI: 10.3390/brainsci12091153] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/30/2022] Open
Abstract
Inflammatory reaction after ischemia-reperfusion contributes significantly to a worsened prognosis, and microglia activation is the main resource of inflammation in the nervous system. Targeting STAT5 has been shown to be a highly effective anti-inflammatory therapy; however, the mechanism by which the STAT5 signaling pathway regulates neuroinflammation following brain injury induced by ischemia-reperfusion remains unclear. Dauricine is an effective agent in anti-inflammation and neuroprotection, but the mechanism by which dauricine acts in ischemia-reperfusion remained unknown. This study is the first to find that the anti-inflammation mechanism of dauricine mainly occurs through the STAT5-NF-κB pathway and that it might act as a STAT5 inhibitor. Dauricine suppresses the inflammation caused by cytokines Eotaxin, KC, TNF-α, IL-1α, IL-1β, IL-6, IL-12β, and IL-17α, as well as inhibiting microglia activation. The STAT5b mutant at Tyr-699 reverses the protective effect of dauricine on the oxygen-glucose deprivation-reperfusion injury of neurons and reactivates the P-NF-κB expression in microglia. These results suggest that dauricine might be able to suppress the neuroinflammation and protect the neurons from the injury of post-ischemia-reperfusion injury via mediating the microglia activation through the STAT5-NF-κB pathway. As a potential therapeutic target for neuroinflammation, STAT5 needs to be given further attention regarding its role in ischemic stroke.
Collapse
|
22
|
Ferulic acid and vinpocetine intake improves memory function by enhancing insulin sensitivity and reducing neuroinflammation and oxidative stress in type 2 diabetic animals with induced Alzheimer's disease. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
23
|
Petraina A, Nogales C, Krahn T, Mucke H, Lüscher TF, Fischmeister R, Kass DA, Burnett JC, Hobbs AJ, Schmidt HHHW. Cyclic GMP modulating drugs in cardiovascular diseases: mechanism-based network pharmacology. Cardiovasc Res 2022; 118:2085-2102. [PMID: 34270705 PMCID: PMC9302891 DOI: 10.1093/cvr/cvab240] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
Mechanism-based therapy centred on the molecular understanding of disease-causing pathways in a given patient is still the exception rather than the rule in medicine, even in cardiology. However, recent successful drug developments centred around the second messenger cyclic guanosine-3'-5'-monophosphate (cGMP), which is regulating a number of cardiovascular disease modulating pathways, are about to provide novel targets for such a personalized cardiovascular therapy. Whether cGMP breakdown is inhibited or cGMP synthesis is stimulated via guanylyl cyclases or their upstream regulators in different cardiovascular disease phenotypes, the outcomes seem to be so far uniformly protective. Thus, a network of cGMP-modulating drugs has evolved that act in a mechanism-based, possibly causal manner in a number of cardiac conditions. What remains a challenge is the detection of cGMPopathy endotypes amongst cardiovascular disease phenotypes. Here, we review the growing clinical relevance of cGMP and provide a glimpse into the future on how drugs interfering with this pathway may change how we treat and diagnose cardiovascular diseases altogether.
Collapse
Affiliation(s)
- Alexandra Petraina
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Cristian Nogales
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Thomas Krahn
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Hermann Mucke
- H.M. Pharma Consultancy, Enenkelstrasse 28/32, A-1160, Vienna, Austria
| | - Thomas F Lüscher
- Royal Brompton & Harefield Hospitals, Heart Division and National Heart and Lung Institute, Guy Scadding Building, Imperial College, Dovehouse Street London SW3 6LY, United Kingdom
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistreet 12, CH-8952 Schlieren, Switzerland
| | - Rodolphe Fischmeister
- INSERM UMR-S 1180, Faculty of Pharmacy, Université Paris-Saclay, F-92296 Châtenay-Malabry, France
| | - David A Kass
- Division of Cardiology, Department of Medicine, Ross Research Building, Rm 858, Johns Hopkins Medical Institutions, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - John C Burnett
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, London, UK
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
24
|
Gu C, Zhang Q, Li Y, Li R, Feng J, Chen W, Ahmed W, Soufiany I, Huang S, Long J, Chen L. The PI3K/AKT Pathway-The Potential Key Mechanisms of Traditional Chinese Medicine for Stroke. Front Med (Lausanne) 2022; 9:900809. [PMID: 35712089 PMCID: PMC9194604 DOI: 10.3389/fmed.2022.900809] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/26/2022] [Indexed: 12/16/2022] Open
Abstract
Stroke is associated with a high disability and fatality rate, and adversely affects the quality of life of patients and their families. Traditional Chinese Medicine (TCM) has been used effectively in the treatment of stroke for more than 2000 years in China and surrounding countries and regions, and over the years, this field has gleaned extensive clinical treatment experience. The Phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT) pathway is important for regulation of cell migration, proliferation, differentiation, and apoptosis, and plays a vital role in vascularization and oxidative stress in stroke. Current Western medicine treatment protocols for stroke include mainly pharmacologic or mechanical thrombectomy to restore blood flow. This review collates recent advances in the past 5 years in the TCM treatment of stroke involving the PI3K/AKT pathway. TCM treatment significantly reduces neuronal damage, inhibits cell apoptosis, and delays progression of stroke via various PI3K/AKT-mediated downstream pathways. In the future, TCM can provide new perspectives and directions for exploring the key factors, and effective activators or inhibitors that affect occurrence and progression of stroke, thereby facilitating treatment.
Collapse
Affiliation(s)
- Chenyang Gu
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Qiankun Zhang
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yajing Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Rong Li
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jia Feng
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wanghao Chen
- Department of Neurosurgery, Shanghai 9th People Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Waqas Ahmed
- School of Medicine, Southeast University, Nanjing, China
| | | | - Shiying Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jun Long
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
25
|
Bian HJ, Xu SY, Li HQ, Jia JQ, Ye L, Shu S, Xia SN, Gu Y, Zhu X, Xu Y, Cao X. JLX001 ameliorates cerebral ischemia injury by modulating microglial polarization and compromising NLRP3 inflammasome activation via the NF-κB signaling pathway. Int Immunopharmacol 2021; 101:108325. [PMID: 34740080 DOI: 10.1016/j.intimp.2021.108325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/15/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is a devastating disease with high morbidity and mortality rates, and the proinflammatory microglia-mediated inflammatory response directly affects stroke outcome. Previous studies have reported that JLX001, a novel compound with a structure similar to that of cyclovirobuxine D (CVB-D), exerts antiapoptotic, anti-inflammatory and antioxidative effects on ischemia-induced brain injury. However, the role of JLX001 in microglial polarization and nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome regulation after ischemic stroke has not been fully investigated. In this study, we used the middle cerebral artery occlusion (MCAO) method to establish a focal cerebral ischemia model and found that JLX001 attenuated the brain infarct size and improved cerebral damage. Moreover, the expression levels of proinflammatory cytokines (interleukin [IL]-1β and tumor necrosis factor [TNF]-α) were significantly reduced while those of the anti-inflammatory cytokine IL-10 were increased in the JLX001-treated group. Immunofluorescence staining and flow cytometry revealed an increased number of anti-inflammatory phenotypic microglia and a reduced number of proinflammatory phenotypic microglia in JLX001-treated MCAO mice. Western blotting analysis showed that JLX001 inhibited the expression of NLRP3 and proteins related to the NLRP3 inflammasome axis in vivo. Furthermore, JLX001 reduced the number of NLRP3/Iba1 cells in ischemic penumbra tissues. Finally, mechanistic analysis revealed that JLX001 significantly inhibited the expression of proteins related to the NF-κB signaling pathway. Additionally, pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor, ameliorated cerebral ischemia-reperfusion injury by suppressing microglial polarization towards the proinflammatory phenotype and NLRP3 activation in vivo, further suggesting that these protective effects of JLX001 were mediated by inhibition of the NF-κB signaling pathway. These results suggest that JLX001 is a promising therapeutic approach for ischemic stroke.
Collapse
Affiliation(s)
- Hui-Jie Bian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Si-Yi Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Hui-Qin Li
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Jun-Qiu Jia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Lei Ye
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Shu Shu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Sheng-Nan Xia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Yue Gu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China
| | - Xiong Zhu
- Jiangsu Jinglixin Pharmaceutical Technology Company Limited, Nanjing 211100, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China.
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China.
| |
Collapse
|
26
|
Acute Inflammation in Cerebrovascular Disease: A Critical Reappraisal with Focus on Human Studies. Life (Basel) 2021; 11:life11101103. [PMID: 34685473 PMCID: PMC8540384 DOI: 10.3390/life11101103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
Recent attention has been focused on the field of inflammatory biomarkers associated with vascular disorders, regarding diagnosis, prognosis, and possible therapeutical targets. In this study, we aimed to perform a comprehensive review of the literature regarding the use of inflammatory biomarkers in stroke patients. We searched studies that evaluated inflammation biomarkers associated with Cerebrovascular Disease (CVD), namely, ischemic Stroke (IS), Intracerebral Hemorrhage (ICH) and Cerebral Venous Thrombosis (CVT). As of today, neutrophil–lymphocyte ratio (NLR) seems the be the most widely studied and accepted biomarker for cerebrovascular disease due to its easy access and availability. Although demonstrated as a prognostic risk factor, in IS, ICH and CVT, its diagnostic role is still under investigation. Several other prognostic factors could be used or even combined together into a diagnostic or prognostic index. Multiple inflammatory biomarkers appear to be involved in IS, ICH, and CVT. Blood inflammatory cells, easily measured and accessible at admission may provide information regarding accurate diagnosis and prognosis. Although not yet a reality, increasing evidence exists to suggest that these may become potential therapeutic targets, likely influencing or mitigating complications of CVD and improving prognosis. Nevertheless, further larger, well-designed randomized clinical trials are still needed to follow up this hypothesis.
Collapse
|
27
|
Yu Z, Jiang N, Su W, Zhuo Y. Necroptosis: A Novel Pathway in Neuroinflammation. Front Pharmacol 2021; 12:701564. [PMID: 34322024 PMCID: PMC8311004 DOI: 10.3389/fphar.2021.701564] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Neuroinflammation is a complex inflammatory process in the nervous system that is expected to play a significant role in neurological diseases. Necroptosis is a kind of necrosis that triggers innate immune responses by rupturing dead cells and releasing intracellular components; it can be caused by Toll-like receptor (TLR)-3 and TLR-4 agonists, tumor necrosis factor (TNF), certain microbial infections, and T cell receptors. Necroptosis signaling is modulated by receptor-interacting protein kinase (RIPK) 1 when the activity of caspase-8 becomes compromised. Activated death receptors (DRs) cause the activation of RIPK1 and the RIPK1 kinase activity-dependent formation of an RIPK1-RIPK3-mixed lineage kinase domain-like protein (MLKL), which is complex II. RIPK3 phosphorylates MLKL, ultimately leading to necrosis through plasma membrane disruption and cell lysis. Current studies suggest that necroptosis is associated with the pathogenesis of neuroinflammatory diseases, such as Alzheimer’s disease, Parkinson’s disease, and traumatic brain injury. Inhibitors of necroptosis, such as necrostatin-1 (Nec-1) and stable variant of Nec (Nec-1s), have been proven to be effective in many neurological diseases. The purpose of this article is to illuminate the mechanism underlying necroptosis and the important role that necroptosis plays in neuroinflammatory diseases. Overall, this article shows a potential therapeutic strategy in which targeting necroptotic factors may improve the pathological changes and clinical symptoms of neuroinflammatory disorders.
Collapse
Affiliation(s)
- Ziyu Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Nan Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.,Department of Pediatric Ophthalmology, Guangzhou Children's Hospital and Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Yao Y, Li Y, Ni W, Li Z, Feng L, Wang Y, Meng J, Zhao H. Systematic Study of Immune Cell Diversity in ischemic postconditioning Using High-Dimensional Single-Cell Analysis with Mass Cytometry. Aging Dis 2021; 12:812-825. [PMID: 34094644 PMCID: PMC8139206 DOI: 10.14336/ad.2020.1115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/15/2020] [Indexed: 11/21/2022] Open
Abstract
Ischemic postconditioning (IPostC) is a concept of ischemic stroke treatment, in which several cycles of brief reocclusion after reperfusion are repeated. It is essential to have an accurate understanding of the immune response in IPostC. By using high parametric single-cell mass cytometry, immune cell subsets and characterize their unique functions from ischemic brain and peripheral blood were identified after IPostC. This study enabled us to better understand the immune cell phenotypical and functional characteristics in ischemic brain and peripheral blood at the single-cell and protein levels. Since some cell surface markers can serve as functional markers, reflecting the degree of inflammation, the cell surface marker intensity among different groups was analyzed. The results showed that downregulation of 4E-BP1 and p38 of Microglia and MoDM in the ischemic brain was involved in IPostC-induced protection. In the peripheral blood, downregulation of P38 of CD4 T cell and Treg has also participated in IPostC-induced protection.
Collapse
Affiliation(s)
- Yang Yao
- 1Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yaning Li
- 1Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Weihua Ni
- 1Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zhijun Li
- 2Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liangshu Feng
- 1Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yan Wang
- 1Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jihong Meng
- 1Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heng Zhao
- 1Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
29
|
Xu H, Wang E, Chen F, Xiao J, Wang M. Neuroprotective Phytochemicals in Experimental Ischemic Stroke: Mechanisms and Potential Clinical Applications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6687386. [PMID: 34007405 PMCID: PMC8102108 DOI: 10.1155/2021/6687386] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/10/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Ischemic stroke is a challenging disease with high mortality and disability rates, causing a great economic and social burden worldwide. During ischemic stroke, ionic imbalance and excitotoxicity, oxidative stress, and inflammation are developed in a relatively certain order, which then activate the cell death pathways directly or indirectly via the promotion of organelle dysfunction. Neuroprotection, a therapy that is aimed at inhibiting this damaging cascade, is therefore an important therapeutic strategy for ischemic stroke. Notably, phytochemicals showed great neuroprotective potential in preclinical research via various strategies including modulation of calcium levels and antiexcitotoxicity, antioxidation, anti-inflammation and BBB protection, mitochondrial protection and antiapoptosis, autophagy/mitophagy regulation, and regulation of neurotrophin release. In this review, we summarize the research works that report the neuroprotective activity of phytochemicals in the past 10 years and discuss the neuroprotective mechanisms and potential clinical applications of 148 phytochemicals that belong to the categories of flavonoids, stilbenoids, other phenols, terpenoids, and alkaloids. Among them, scutellarin, pinocembrin, puerarin, hydroxysafflor yellow A, salvianolic acids, rosmarinic acid, borneol, bilobalide, ginkgolides, ginsenoside Rd, and vinpocetine show great potential in clinical ischemic stroke treatment. This review will serve as a powerful reference for the screening of phytochemicals with potential clinical applications in ischemic stroke or the synthesis of new neuroprotective agents that take phytochemicals as leading compounds.
Collapse
Affiliation(s)
- Hui Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 508060, China
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | | | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 508060, China
| | - Jianbo Xiao
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Mingfu Wang
- Institute for Advanced Study, Shenzhen University, Shenzhen 508060, China
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| |
Collapse
|
30
|
Song Z, Feng J, Zhang Q, Deng S, Yu D, Zhang Y, Li T. Tanshinone IIA Protects Against Cerebral Ischemia Reperfusion Injury by Regulating Microglial Activation and Polarization via NF-κB Pathway. Front Pharmacol 2021; 12:641848. [PMID: 33953677 PMCID: PMC8090935 DOI: 10.3389/fphar.2021.641848] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
Tanshinone IIA, a fat-soluble diterpenoid isolated from Salvia miltiorrhiza Bunge, has been shown to attenuate the cerebral ischemic injury. The aim of this study was to examine the effects on neuroprotection and microglia activation of Tanshinone IIA. Male Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO). We found that Tanshinone IIA significantly reduced infarction volume, alleviated neuronal injuries, reduced the release of TNF-α, IL-1β, and IL-6, increased SOD activity, and decrease the content of MDA in MCAO rats. Hematoxylin and eosin staining, Nissl staining, TUNEL staining and immunofluorescence staining showed that Tanshinone IIA improved the distribution and morphology of neurons in brain tissues and reduced apoptosis. In addition, Co-immunofluorescence staining of rat brain tissues and the mRNA expression levels of CD11b, CD32, iNOS, and Arg-1, CD206, IL-10 in BV2 cells indicated that Tanshinone IIA can downregulate M1 microglia and upregulate M2 microglia in MCAO rats. Further, BV2 microglial cells were subjected to oxygen-glucose deprivation, the protein expression levels were detected by western blot. Tanshinone IIA inhibited the expression levels of NF-κB signaling pathway related proteins. Taken together, this study suggested that Tanshinone IIA modulated microglial M1/M2 polarization via the NF-κB signaling pathway to confer anti-neuroinflammatory effects.
Collapse
Affiliation(s)
- Zhibing Song
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China.,College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Jingjing Feng
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Qian Zhang
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China
| | - Shanshan Deng
- School of Medicine, Shanghai University, Shanghai, China
| | | | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, China
| | - Tiejun Li
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China
| |
Collapse
|
31
|
Zang J, Wu Y, Su X, Zhang T, Tang X, Ma D, Li Y, Liu Y, Weng Z, Liu X, Tsang CK, Xu A, Lu D. Inhibition of PDE1-B by Vinpocetine Regulates Microglial Exosomes and Polarization Through Enhancing Autophagic Flux for Neuroprotection Against Ischemic Stroke. Front Cell Dev Biol 2021; 8:616590. [PMID: 33614626 PMCID: PMC7889976 DOI: 10.3389/fcell.2020.616590] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/31/2020] [Indexed: 12/17/2022] Open
Abstract
Exosomes contribute to cell–cell communications. Emerging evidence has shown that microglial exosomes may play crucial role in regulation of neuronal functions under ischemic conditions. However, the underlying mechanisms of microglia-derived exosome biosynthesis are largely unknown. Herein, we reported that the microglial PDE1-B expression was progressively elevated in the peri-infarct region after focal middle cerebral artery occlusion. By an oxygen-glucose-deprivation (OGD) ischemic model in cells, we found that inhibition of PDE1-B by vinpocetine in the microglial cells promoted M2 and inhibited M1 phenotype. In addition, knockdown or inhibition of PDE1-B significantly enhanced the autophagic flux in BV2 cells, and vinpocetine-mediated suppression of M1 phenotype was dependent on autophagy in ischemic conditions. Co-culture of BV2 cells and neurons revealed that vinpocetine-treated BV2 cells alleviated OGD-induced neuronal damage, and treatment of BV2 cells with 3-MA abolished the observed effects of vinpocetine. We further demonstrated that ischemia and vinpocetine treatment significantly altered microglial exosome biogenesis and release, which could be taken up by recipient neurons and regulated neuronal damage. Finally, we showed that the isolated exosome per se from conditioned BV2 cells is sufficient to regulate cortical neuronal survival in vivo. Taken together, these results revealed a novel microglia-neuron interaction mediated by microglia-derived exosomes under ischemic conditions. Our findings further suggest that PDE1-B regulates autophagic flux and exosome biogenesis in microglia which plays a crucial role in neuronal survival under cerebral ischemic conditions.
Collapse
Affiliation(s)
- Jiankun Zang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yousheng Wu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xuanlin Su
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tianyuan Zhang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xionglin Tang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Dan Ma
- Section of Molecular Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| | - Yufeng Li
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yanfang Liu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ze'an Weng
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xuanzhuo Liu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chi Kwan Tsang
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Anding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dan Lu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
32
|
Zhuang S, Liu B, Guo S, Xue Y, Wu L, Liu S, Zhang C, Ni X. Germacrone alleviates neurological deficits following traumatic brain injury by modulating neuroinflammation and oxidative stress. BMC Complement Med Ther 2021; 21:6. [PMID: 33402180 PMCID: PMC7786997 DOI: 10.1186/s12906-020-03175-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/03/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Germacrone (GM) is a terpenoid compound which is reported to have anti-inflammatory and anti-oxidative effects. However, its role in treating traumatic brain injury (TBI) remains largely unknown. METHODS Male C57BL/6 mice were divided into the following groups: control group, TBI group [controlled cortical impact (CCI) model], CCI + 5 mg/kg GM group, CCI + 10 mg/kg GM group and CCI + 20 mg/kg GM group. GM was administered via intraperitoneal injection. The neurological functions (including motor coordination, spatial learning and memory abilities) and brain edema were measured. Nissl staining was used to detect the neuronal apoptosis. Colorimetric assays and enzyme linked immunosorbent assay (ELISA) kits were used to determine the expression levels of oxidative stress markers including myeloperoxidase (MPO), malondialdehyde (MDA) and superoxide dismutase (SOD), as well as the expressions of inflammatory markers, including tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6). Additionally, protein levels of Nrf2 and p-p65 were detected by Western blot assay. RESULTS GM significantly ameliorated motor dysfunction, spatial learning and memory deficits of the mice induced by TBI and it also reduced neuronal apoptosis and microglial activation in a dose-dependent manner. Besides, GM treatment reduced neuroinflammation and oxidative stress compared to those in the CCI group in a dose-dependent manner. Furthermore, GM up-regulated the expression of antioxidant protein Nrf2 and inhibited the expression of inflammatory response protein p-p65. CONCLUSIONS GM is a promising drug to improve the functional recovery after TBI via repressing neuroinflammation and oxidative stress.
Collapse
Affiliation(s)
- Sujing Zhuang
- Department of Neurology, Linyi Central Hospital, Linyi, 276400, Shandong, China
| | - Baogui Liu
- Department of Anesthesiology, Linyi Central Hospital, Linyi, 276400, Shandong, China
| | - Shifeng Guo
- Department of Neurology, Linyi Central Hospital, Linyi, 276400, Shandong, China
| | - Yanzhong Xue
- Department of Neurology, Linyi Central Hospital, Linyi, 276400, Shandong, China
| | - Lin Wu
- Department of Intensive Care Unit 2, Affiliated Hospital of Jining Medical University, Jining, 272000, Shandong, China
| | - Shiqi Liu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jining Medical University, Jining, 272000, Shandong, China
| | - Chunling Zhang
- Department of Intensive Care Unit 2, Affiliated Hospital of Jining Medical University, Jining, 272000, Shandong, China
| | - Xiuyan Ni
- Department of Radiotherapy, Linyi Central Hospital, Linyi, 276400, Shandong, China.
| |
Collapse
|
33
|
Han D, Wang J, Wen L, Sun M, Liu H, Gao Y. Vinpocetine Attenuates Ischemic Stroke Through Inhibiting NLRP3 Inflammasome Expression in Mice. J Cardiovasc Pharmacol 2020; 77:208-216. [PMID: 33351536 PMCID: PMC7853762 DOI: 10.1097/fjc.0000000000000945] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/17/2020] [Indexed: 11/25/2022]
Abstract
ABSTRACT Ischemic stroke is the leading cause of globe death and permanent disability, but its therapeutic strategies are limited. Over the past decades, multiprotein complexes called inflammasomes have been shown as promising targets in ischemic stroke. Here, we examined vinpocetine (Vinp), a synthetic drug, playing a neuroprotective role against ischemic stroke in mice through regulating NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation. Middle cerebral artery occlusion/reperfusion (MCAO/R) was applied to mimic ischemic stroke in vivo. Vinp was administrated by intraperitoneal injection with different dose (5 or 10 mg/kg) 1 hour after reperfusion. Then, neurological assessment and infarct size were performed, and interleukin-1β (IL-1β) and IL-18 levels were evaluated using ELISA. The levels of NLRP3 inflammasome components and its upstream nuclear factor-κB (NF-κB) were determined using real-time PCR or Western blot. The experimental results indicated that posttreatment with Vinp decreased cerebral infarct size, improved behavior recover, reduced NLRP3 inflammasome expression, and suppressed the transfer of NF-κB to nucleus and proinflammatory cytokine release in middle cerebral artery occlusion/reperfusion mice. In conclusion, this study demonstrates that Vinp alleviates ischemic stroke by regulating levels of NLRP3 inflammasome, NF-κB, and proinflammatory cytokines in vivo, offering an alternative medication for ischemic stroke associated with inflammation.
Collapse
Affiliation(s)
- Dong Han
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Peopleʹs Republic of China.
| | - Jue Wang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Peopleʹs Republic of China.
| | - Lulu Wen
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Peopleʹs Republic of China.
| | - Miao Sun
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Peopleʹs Republic of China.
| | - Hang Liu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Peopleʹs Republic of China.
| | - Yan Gao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Peopleʹs Republic of China.
| |
Collapse
|
34
|
Zhou Q, Guo D, Li X, Wang Y, Ye X, Xue S, Wang X. Anti-inflammatory effects of vinpocetine in LPS-stimulated microglia via activation of AMPK. AN ACAD BRAS CIENC 2020; 92:e20200241. [PMID: 33237143 DOI: 10.1590/0001-3765202020200241] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/17/2020] [Indexed: 11/21/2022] Open
Abstract
Microglia are the resident immune cells in the central nervous system (CNS), which play important roles in the repair of neuroinflammatory injury. The present study investigated the anti-neuroinflammatory effects of vinpocetine induced by lipopolysaccharide (LPS) in BV2 microglia. BV2 microglia were pretreated with vinpocetine, and then stimulated with LPS (100 ng/mL). The cytotoxicity of BV2 microglia was assessed by MTT assay. The expression levels of nitrite oxide were measured by Griess assay. Proinflammatory cytokines and mediators were determined by Western blot, ELISA, or quantitative real-time PCR. Vinpocetine significantly decreased the generation of nitric oxide-inducible nitric oxide synthase (iNOS), cyclooxygenase- (COX-) 2 in a dose-dependent manner. In addition, vinpocetine decreased the production of pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α), interleukin (IL)-6 and IL-1β. Furthermore, it was observed that phosphorylation levels of AMPK (Thr-172) decreased in LPS-stimulated BV2 microglia. Vinpocetine treatment increased AMPK phosphorylation in LPS-stimulated BV2 microglia. AMPK inhibition by siRNA blocked the anti-inflammatory effects of vinpocetine induced by LPS in BV2 microglia. The overall results demonstrate that vinpocetine has anti-inflammatory effects on LPS-stimulated BV2 microglia via inducing phosphorylation of AMPK, suggesting that vinpocetine is a potential therapeutic agent in neuroinflammatory injury.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, 215002, P.R. China
| | - Dongkai Guo
- Department of pharmacy, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215153, P.R. China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases Research and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215021, P.R. China
| | - Xinjia Li
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, 215002, P.R. China
| | - Yixuan Wang
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, 215002, P.R. China
| | - Xiaoli Ye
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, 215002, P.R. China
| | - Sudong Xue
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, 215002, P.R. China
| | - Xiaoyu Wang
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, 215002, P.R. China
| |
Collapse
|
35
|
Yıldırım E, Sezer G. Clinical plasma concentration of vinpocetine does not affect osteogenic differentiation of mesenchymal stem cells. Pharmacol Rep 2020; 73:202-210. [PMID: 32865810 DOI: 10.1007/s43440-020-00153-8] [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: 05/12/2020] [Revised: 07/17/2020] [Accepted: 08/20/2020] [Indexed: 11/30/2022]
Abstract
AIM Vinpocetine (Vin) has long been used as a medicine to treat cerebrovascular disorders and as a dietary supplement to improve cognitive functions. Previous studies have revealed that the transcription factor nuclear factor kappa B (NF-κB) activity plays an important role in osteogenic differentiation of mesenchymal stem cells (MSC). Vin inhibits NF-κB-dependent inflammatory responses; however, the effect of Vin on the osteogenic differentiation of MSCs has not been reported. In this study, we aimed to the investigate effect of Vin on the osteogenic differentiation of rat bone marrow-derived MSCs (BMSCs). METHODS We treated BMSCs with clinical plasma (0.17 µM) or higher concentrations (5 and 20 µM) of Vin with no significant effect on the cell viability. Alizarin Red S and alkaline phosphatase (ALP) stainings were used to evaluate mineralizations on days 14 and 21. Moreover, expressions of target genes were detected using qRT-PCR analysis. RESULTS Osteogenic differentiation of BMSCs did not significantly change with Vin's clinical plasma concentration, but significantly decreased with higher concentrations. Calcium mineralization, ALP staining and mRNA gene expressions of Runx2 and ALP were decreased significantly with high concentrations of Vin, paticularly on day 21. CONCLUSION Our in vitro findings suggest that clinically relevant concentration of Vin seems safe to use in elderly patients with respect to osteoporosis. On the other hand, Vin at high concentrations appears to be harmful to bone homeostasis.
Collapse
Affiliation(s)
- Esma Yıldırım
- Pharmacy Division, Ministry of Health Kayseri City Hospital, 38080, Kayseri, Turkey
| | - Gulay Sezer
- School of Medicine, Pharmacology Department, Erciyes University, 38039, Kayseri, Turkey. .,Genkok Genome and Stem Cell Centre, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
36
|
Liu S, Liu P, Wang P, Zhang F, Wang L, Wang Y, Lu H, Ma X. Argatroban Increased the Basal Vein Drainage and Improved Outcomes in Acute Paraventricular Ischemic Stroke Patients. Med Sci Monit 2020; 26:e924593. [PMID: 32667287 PMCID: PMC7382300 DOI: 10.12659/msm.924593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Since venous drainage in acute arterial ischemic stroke has not been thoroughly researched, we evaluate the effect of argatroban, a selective direct thrombin inhibitor, as a therapy to increase the rate of basal vein Rosenthal (BVR) drainage and improve patients’ post-stroke outcomes. Material/Methods In this multicenter clinical trial, 60 eligible patients at 4.5 to 48 hours after the stroke onset were recruited. After being randomly allocated into 2 groups, they were treated with standard therapy either alone or with argatroban. Results Compared to the contralateral brain hemisphere, the mean flow velocity (MFV) in BVR drainage was significantly reduced in the stroke-afflicted ipsilateral hemisphere. After treatment with argatroban for 7 days, the MFV from BVR of the ipsilateral hemisphere in the argatroban treated group was significantly increased when compared to the control group. At 90 days after the onset of stroke, the MFV of BVR in the ipsilateral hemisphere was similar in both groups. Compared with controls, the argatroban-treated patients had smaller lesions from baseline to 7 days. Argatroban also improved National Institutes of Health Stroke Scale (NIHSS) scores on day 7 after the onset of stroke. Furthermore, the argatroban group’s neurological functions were superior to those of their untreated counterparts after 90 days. No difference was found in the incidence of adverse reactions between the 2 groups. Conclusions These observations indicate that vein drainage change may contribute to the acute phase of brain edema and the outcomes of ischemic stroke patients. Clinical Trial Registration URL-http://www.chictr.org Unique identifier: ChiCTR-IPR-16008663
Collapse
Affiliation(s)
- Shoufeng Liu
- The Graduate School, Tianjin Medical University, Tianjin, China (mainland).,Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China (mainland)
| | - Peipei Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China (mainland)
| | - Po Wang
- Department of Neurology, Baotou Central Hospital, Baotou, Inner Mongolia, China (mainland)
| | - Fang Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Lijun Wang
- Department of Neurology, Tianjin Fourth Central Hospital, Tianjin, China (mainland)
| | - Yu Wang
- Department of Ultrasonography, Tianjin Huanhu Hospital, Tianjin, China (mainland)
| | - Hao Lu
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China (mainland)
| | - Xiaofeng Ma
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| |
Collapse
|
37
|
Sanders O, Rajagopal L. Phosphodiesterase Inhibitors for Alzheimer's Disease: A Systematic Review of Clinical Trials and Epidemiology with a Mechanistic Rationale. J Alzheimers Dis Rep 2020; 4:185-215. [PMID: 32715279 PMCID: PMC7369141 DOI: 10.3233/adr-200191] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Preclinical studies, clinical trials, and reviews suggest increasing 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) with phosphodiesterase inhibitors is disease-modifying in Alzheimer's disease (AD). cAMP/protein kinase A (PKA) and cGMP/protein kinase G (PKG) signaling are disrupted in AD. cAMP/PKA and cGMP/PKG activate cAMP response element binding protein (CREB). CREB binds mitochondrial and nuclear DNA, inducing synaptogenesis, memory, and neuronal survival gene (e.g., brain-derived neurotrophic factor) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α). cAMP/PKA and cGMP/PKG activate Sirtuin-1, which activates PGC1α. PGC1α induces mitochondrial biogenesis and antioxidant genes (e.g.,Nrf2) and represses BACE1. cAMP and cGMP inhibit BACE1-inducing NFκB and tau-phosphorylating GSK3β. OBJECTIVE AND METHODS We review efficacy-testing clinical trials, epidemiology, and meta-analyses to critically investigate whether phosphodiesteraseinhibitors prevent or treat AD. RESULTS Caffeine and cilostazol may lower AD risk. Denbufylline and sildenafil clinical trials are promising but preliminary and inconclusive. PF-04447943 and BI 409,306 are ineffective. Vinpocetine, cilostazol, and nicergoline trials are mixed. Deprenyl/selegiline trials show only short-term benefits. Broad-spectrum phosphodiesterase inhibitor propentofylline has been shown in five phase III trials to improve cognition, dementia severity, activities of daily living, and global assessment in mild-to-moderate AD patients on multiple scales, including the ADAS-Cogand the CIBIC-Plus in an 18-month phase III clinical trial. However, two books claimed based on a MedScape article an 18-month phase III trial failed, so propentofylline was discontinued. Now, propentofylline is used to treat canine cognitive dysfunction, which, like AD, involves age-associated wild-type Aβ deposition. CONCLUSION Phosphodiesterase inhibitors may prevent and treat AD.
Collapse
|
38
|
Huang JL, Liu WW, Manaenko A, Sun XJ, Mei QY, Hu Q. Hydrogen inhibits microglial activation and regulates microglial phenotype in a mouse middle cerebral artery occlusion model. Med Gas Res 2020; 9:127-132. [PMID: 31552875 PMCID: PMC6779010 DOI: 10.4103/2045-9912.266987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Microglia participate in bi-directional control of brain repair after stroke. Previous studies have demonstrated that hydrogen protects brain after ischemia/reperfusion (I/R) by inhibiting inflammation, but the specific mechanism of anti-inflammatory effect of hydrogen is poorly understood. The goal of our study is to investigate whether inhalation of high concentration hydrogen (HCH) is able to attenuate I/R-induced microglia activation. Eighty C57B/L male mice were divided into four groups: sham, I/R, I/R + HCH and I/R + N2/O2 groups. Assessment of animals happened in “blind” matter. I/R was induced by occlusion of middle cerebral artery for one hour). After one hour, filament was withdrawn, which induced reperfusion. Hydrogen treated I/R animals inhaled mix of 66.7% H2 balanced with O2 for 90 minutes, starting immediately after initiation of reperfusion. Control animals (N2/O2) inhaled mix in which hydrogen was replaced with N2 for the same time (90 minutes). The brain injury, such as brain infarction and development of brain edema, as well as neurobehavioral deficits were determined 23 hours after reperfusion. Effect of HCH on microglia activation in the ischemic penumbra was investigated by immunostaining also 23 hours after reperfusion. mRNA expression of inflammation related genes was detected by PCR. Our results showed that HCH attenuated brain injury and consequently reduced neurological dysfunction after I/R. Furthermore, we demonstrated that HCH directed microglia polarization towards anti-inflammatory M2 polarization. This study indicates hydrogen may exert neuroprotective effects by inhibiting the microglial activation and regulating microglial polarization. This study was conducted in agreement with the Animal Care and Use Committee (IACUC) and Institutional Animal Care guidelines regulation (Shanghai Jiao Tong University, China (approval No. A2015-011) in November 2015.
Collapse
Affiliation(s)
- Jun-Long Huang
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine; Department of Navy Aviation Medicine, Faculty of Naval Medicine, the Naval Military Medical University; Department of Navy Aviation Medicine, Naval Medical center of PLA, the Naval Military Medical University, Shanghai, China
| | - Wen-Wu Liu
- Department of Diving Medicine, Faculty of Naval Medicine, the Naval Military Medical University, Shanghai, China
| | - Anatol Manaenko
- Departments of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Xue-Jun Sun
- Department of Navy Aviation Medicine, Faculty of Naval Medicine, the Naval Military Medical University, Shanghai, China
| | - Qi-Yong Mei
- Department of Neurosurgery, Changzheng Hospital, the Naval Military Medical University, Shanghai, China
| | - Qin Hu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Yang XL, Cao CZ, Zhang QX. MiR-195 alleviates oxygen-glucose deprivation/reperfusion-induced cell apoptosis via inhibition of IKKα-mediated NF-κB pathway. Int J Neurosci 2020; 131:755-764. [PMID: 32271641 DOI: 10.1080/00207454.2020.1754212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Increasing evidence confirmed that miRNA plays a critical role in the occurrence and development of ischemic stroke. Here, the aim of this study was to examine the function and mechanisms of miR-195 in vascular endothelial cell apoptosis induced by oxygen-glucose deprivation (OGD). METHODS This study intended to use OGD to simulate ischemia in vitro. The mRNA expression of miR-195, IKKα and NF-κB in human umbilical vein endothelial cells (HUVECs) were detected by RT-qPCR. The proliferation and apoptosis ability of HUVECs were evaluated using MTT assay, colony formation assay and flow cytometry, respectively. Western blot was applied to examine related protein expression. The interaction between miR-195 and IKKα was verified by dual-luciferase reporter gene assay. RESULTS OGD significantly inhibited cell viability and induced cell apoptosis in HUVECs. Meanwhile, OGD treatment notably decreased the expression of miR-195, as well as enhanced NF-κB expression. Moreover, miR-195 directly interacted with IKKα and suppressed its expression. Mechanically, overexpression of miR-195 exhibited pro-proliferation and anti-apoptotic effect on HUVECs treated with OGD through targeting IKKα-mediated NF-κB pathway. At the molecular level, through suppressing IKKα/NF-κB pathway, miR-195 inhibited the expression of pro-apoptotic protein Bax and active caspase-3, but increased the expression of anti-apoptotic Bcl-2 in HUVECs. CONCLUSIONS Our finding uncovers the protective effect of miR-195 on the biological behavior of HUVECs via suppression of the NF-κB pathway induced by IKKα, which may provide a new potential strategy for ischemic stroke clinical treatment.
Collapse
Affiliation(s)
- Xiao-Li Yang
- Department of Neurology, Qinghai Provincial People's Hospital, Xining, P. R. China
| | - Cheng-Zhu Cao
- Department of Physiology, Medical College of Qinghai University, Xining, P. R. China
| | - Qing-Xin Zhang
- Department of Radiology, Qinghai Provincial People's Hospital, Xining, P. R. China
| |
Collapse
|
40
|
Zhao M, Hou S, Feng L, Shen P, Nan D, Zhang Y, Wang F, Ma D, Feng J. Vinpocetine Protects Against Cerebral Ischemia-Reperfusion Injury by Targeting Astrocytic Connexin43 via the PI3K/AKT Signaling Pathway. Front Neurosci 2020; 14:223. [PMID: 32300287 PMCID: PMC7142276 DOI: 10.3389/fnins.2020.00223] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/28/2020] [Indexed: 12/21/2022] Open
Abstract
Vinpocetine (Vinp) is known for its neuroprotective properties. However, the protective mechanism of Vinp against cerebral ischemia/reperfusion (I/R) injury should be further explored. This study was designed to investigate the neuroprotective effects of Vinp against oxygen-glucose deprivation/reoxygenation (OGD/R) injury in vitro and cerebral I/R injury in vivo and explore whether this mechanism would involve enhancement of astrocytic connexin 43 (Cx43) expression via the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway. In vitro, we detected astrocytic viability and extracellular nitric oxide by an assay kit, intracellular reactive oxygen species by a DCFH-DA probe, inflammation and apoptosis-related protein expression by immunofluorescence staining, and the astrocytic apoptosis rate by flow cytometry. In vivo, we measured the cerebral infarction volume, superoxide dismutase activity, malondialdehyde content, and the expression of inflammation and apoptosis-related proteins. The results indicated that Vinp ameliorated the detrimental outcome of I/R injury. Vinp attenuated astrocytic injury induced by OGD/R and reduced cerebral infarction volume and cerebral edema in rats with cerebral I/R injury. Moreover, Vinp reduced oxidative stress, inflammation, and apoptosis induced by cerebral I/R injury in brain tissues. Meanwhile, Vinp increased p-Cx43 and p-AKT expression, and the p-Cx43/Cx43 and p-AKT/AKT ratio, which was decreased by cerebral I/R injury. Coadministration of PI3K inhibitors LY294002 and BKM120 blunted the effects of Vinp. This study suggests that Vinp protects against cerebral I/R injury via Cx43 phosphorylation by activating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Shuai Hou
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Liangshu Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Pingping Shen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Di Nan
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yunhai Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China.,Jiangsu Key Laboratory of Medical Optics, Suzhou, China
| | - Famin Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China.,Jiangsu Key Laboratory of Medical Optics, Suzhou, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
41
|
Rashid MH, Kabir A, Waris MU, Salman U, Zain S. Role of Prophylactic Antibiotics in Critical Care of Stroke Patients - A Preventive Approach to Post-stroke Infections? Cureus 2020; 12:e7158. [PMID: 32257701 PMCID: PMC7108674 DOI: 10.7759/cureus.7158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Post-stroke complications are very common worldwide and the most common complication is infection. This contributes the most to the mortality rate in stroke patients. Among the infections, pneumonia and urinary tract infections are most common. Hyperthermia following stroke is associated with neuronal damage and worse outcomes. Post-stroke immunosuppression and activation of inflammatory mediators also cause infections. Based on the high mortality caused by post-stroke infections, various trials were done to seek the advantage that prophylactic antibiotics can give in the critical care of stroke patients. Antibiotics, including ceftriaxone (cephalosporin), levofloxacin (fluoroquinolone), penicillin, and minocycline (tetracycline), were used and the stroke patients were followed up to analyze the primary and secondary outcomes. It was concluded that early antibiotic therapy (mostly within 24 hours) leads to a reduced rate of post-stroke infections and reduced fever spikes, whereas follow-up for a longer period of time showed no better functional outcome. Furthermore, mortality and morbidity benefits were also not seen with prophylactic antibiotic therapy. This review helped us to put a nail in the coffin to the earlier thoughts that prophylactic antibiotics are necessary for the critical care of stroke patients.
Collapse
Affiliation(s)
- Muhammad Humayoun Rashid
- Neurology, Bakhtawar Amin Medical and Dental College, Multan, PAK.,Internal Medicine, Nishtar Medical University and Hospital, Multan, PAK
| | - Ahmad Kabir
- Pathology, Bakhtawar Amin Medical and Dental College, Multan, PAK.,Internal Medicine, Nishtar Hospital, Multan, PAK
| | | | - Umer Salman
- Internal Medicine, City Hospital, Multan, PAK
| | - Sarmad Zain
- Internal Medicine, Nishtar Hospital, Nishtar Medical University, Multan, PAK
| |
Collapse
|
42
|
Al-Kuraishy HM, Al-Gareeb AI, Naji MT, Al-Mamorry F. Role of vinpocetine in ischemic stroke and poststroke outcomes: A critical review. Brain Circ 2020; 6:1-10. [PMID: 32166194 PMCID: PMC7045535 DOI: 10.4103/bc.bc_46_19] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/06/2019] [Accepted: 11/18/2019] [Indexed: 02/06/2023] Open
Abstract
Vinpocetine (VPN) is a synthetic ethyl-ester derivative of the alkaloid apovincamine from Vinca minor leaves. VPN is a selective inhibitor of phosphodiesterase type 1 (PDE1) that has potential neurological effects through inhibition of voltage-gated sodium channel and reduction of neuronal calcium influx. VPN has noteworthy antioxidant, anti-inflammatory, and anti-apoptotic effects with inhibitory effect on glial and astrocyte cells during and following ischemic stroke (IS). VPN is effective as adjuvant therapy in the management of epilepsy; it reduces seizure frequency by 50% in a dose of 2 mg/kg/day. VPN improves psychomotor performances through modulation of brain monoamine pathway mainly on dopamine and serotonin, which play an integral role in attenuation of depressive symptoms. VPN recover cognitive functions and spatial memory through inhibition of hippocampal and cortical PDE1 with augmentation of cyclic adenosin monophosphate and cyclic guanosin monophosphate ratio, enhancement of cholinergic neurotransmission, and inhibition of neuronal inflammatory mediators. Therefore, VPN is an effective agent in the management of IS and plays an integral role in the prevention and attenuation of poststroke epilepsy, depression, and cognitive deficit through direct cAMP/cGMP-dependent pathway or indirectly through anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Hayder M. Al-Kuraishy
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Almustansiriya University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Almustansiriya University, Baghdad, Iraq
| | - Marwa Thaier Naji
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Almustansiriya University, Baghdad, Iraq
| | - Farah Al-Mamorry
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Almustansiriya University, Baghdad, Iraq
| |
Collapse
|
43
|
The Role of Oxidative Stress in Common Risk Factors and Mechanisms of Cardio-Cerebrovascular Ischemia and Depression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2491927. [PMID: 32148646 PMCID: PMC7044480 DOI: 10.1155/2019/2491927] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/01/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The public health sector faces a huge challenge as a result of the high prevalence and burden of disability caused by ischemic cardio-cerebrovascular disease (CVD) and depression. Although studies have explored the underlying mechanisms and potential therapies to address conditions, there is no treatment breakthrough, especially for depression which is highly influenced by social stressors. However, accumulating evidence reveals that CVD and depression are correlated and share common risk factors, particularly obesity, diabetes, and hypertension. They also share common mechanisms, including oxidative stress (OS), inflammation and immune response, cell death signaling pathway, and microbiome-gut-brain axis. This review summarizes the relationship between ischemic CVD and depression and describes the interactions among common risk factors and mechanisms for these two diseases. In addition, we propose that OS mediates the crosstalk between these diseases. We also reveal the potential of antioxidants to ameliorate OS-related injuries.
Collapse
|
44
|
Nouri Z, Fakhri S, El-Senduny FF, Sanadgol N, Abd-ElGhani GE, Farzaei MH, Chen JT. On the Neuroprotective Effects of Naringenin: Pharmacological Targets, Signaling Pathways, Molecular Mechanisms, and Clinical Perspective. Biomolecules 2019; 9:E690. [PMID: 31684142 PMCID: PMC6920995 DOI: 10.3390/biom9110690] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022] Open
Abstract
As a group of progressive, chronic, and disabling disorders, neurodegenerative diseases (NDs) affect millions of people worldwide, and are on the rise. NDs are known as the gradual loss of neurons; however, their pathophysiological mechanisms have not been precisely revealed. Due to the complex pathophysiological mechanisms behind the neurodegeneration, investigating effective and multi-target treatments has remained a clinical challenge. Besides, appropriate neuroprotective agents are still lacking, which raises the need for new therapeutic agents. In recent years, several reports have introduced naturally-derived compounds as promising alternative treatments for NDs. Among natural entities, flavonoids are multi-target alternatives affecting different pathogenesis mechanisms in neurodegeneration. Naringenin is a natural flavonoid possessing neuroprotective activities. Increasing evidence has attained special attention on the variety of therapeutic targets along with complex signaling pathways for naringenin, which suggest its possible therapeutic applications in several NDs. Here, in this review, the neuroprotective effects of naringenin, as well as its related pharmacological targets, signaling pathways, molecular mechanisms, and clinical perspective, are described. Moreover, the need to develop novel naringenin delivery systems is also discussed to solve its widespread pharmacokinetic limitation.
Collapse
Affiliation(s)
- Zeinab Nouri
- Student's Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran.
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Fardous F El-Senduny
- Biochemistry division, Chemistry Department, Faculty of Science, Mansoura University, 35516 Mansoura, Egypt.
| | - Nima Sanadgol
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol 7383198616, Iran.
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto 14040-903, Brazil.
| | - Ghada E Abd-ElGhani
- Department of Chemistry, Faculty of Science, University of Mansoura, 35516 Mansoura, Egypt.
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Jen-Tsung Chen
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung 811, Taiwan.
| |
Collapse
|
45
|
Baillie GS, Tejeda GS, Kelly MP. Therapeutic targeting of 3',5'-cyclic nucleotide phosphodiesterases: inhibition and beyond. Nat Rev Drug Discov 2019; 18:770-796. [PMID: 31388135 PMCID: PMC6773486 DOI: 10.1038/s41573-019-0033-4] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2019] [Indexed: 01/24/2023]
Abstract
Phosphodiesterases (PDEs), enzymes that degrade 3',5'-cyclic nucleotides, are being pursued as therapeutic targets for several diseases, including those affecting the nervous system, the cardiovascular system, fertility, immunity, cancer and metabolism. Clinical development programmes have focused exclusively on catalytic inhibition, which continues to be a strong focus of ongoing drug discovery efforts. However, emerging evidence supports novel strategies to therapeutically target PDE function, including enhancing catalytic activity, normalizing altered compartmentalization and modulating post-translational modifications, as well as the potential use of PDEs as disease biomarkers. Importantly, a more refined appreciation of the intramolecular mechanisms regulating PDE function and trafficking is emerging, making these pioneering drug discovery efforts tractable.
Collapse
Affiliation(s)
- George S Baillie
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Gonzalo S Tejeda
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| |
Collapse
|
46
|
Baram SM, Karima S, Shateri S, Tafakhori A, Fotouhi A, Lima BS, Rajaei S, Mahdavi M, Tehrani HS, Aghamollaii V, Aghamiri SH, Mansouri B, Gharahje S, Kabiri S, Hosseinizadeh M, Shahamati SZ, Alborzi AT. Functional improvement and immune-inflammatory cytokines profile of ischaemic stroke patients after treatment with boswellic acids: a randomized, double-blind, placebo-controlled, pilot trial. Inflammopharmacology 2019; 27:1101-1112. [DOI: 10.1007/s10787-019-00627-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022]
|
47
|
Liu PY, Zhang Z, Liu Y, Tang XL, Shu S, Bao XY, Zhang Y, Gu Y, Xu Y, Cao X. TMEM16A Inhibition Preserves Blood-Brain Barrier Integrity After Ischemic Stroke. Front Cell Neurosci 2019; 13:360. [PMID: 31447648 PMCID: PMC6691060 DOI: 10.3389/fncel.2019.00360] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/23/2019] [Indexed: 12/23/2022] Open
Abstract
The inflammatory response plays a pivotal role in Blood–Brain Barrier (BBB) destruction following ischemic brain injury. Enhanced leukocyte adhesion to vascular endothelial cells is an essential event in the inflammatory process. TMEM16A, a newly discovered protein regulating calcium-activated chloride channels, is widely expressed in eukaryotes. Recent studies have suggested that upregulated expression of TMEM16A is associated with the occurrence and development of many diseases. However, the role of TMEM16A in regulating BBB integrity after ischemic stroke has not been fully investigated. In this study, we found that TMEM16A is mainly expressed in brain endothelial cells and upregulated after ischemic stroke in the mouse brain. Caccinh-A01, an TMEM16A inhibitor that reduced its upregulation, attenuated brain infarct size and neurological deficits after ischemic stroke. ICAM-1 and MPO expression and BBB permeability were decreased after TMEM16A inhibitor administration. In addition, TMEM16A silencing rescued oxygen-glucose deprivation/reoxygenation (OGD/R)-induced transendothelial permeability in vitro accompanied by decreased ICAM-1 expression and leukocyte adhesion. Furthermore, our mechanistic study showed that TMEM16A knockdown alleviated NF-κB activation and nuclear translocation, indicating that TMEM16A knockdown downregulated OGD/R-induced ICAM-1 expression in an NF-κB-dependent manner. Finally, NF-κB inhibitor treatment also alleviated OGD/ R-induced BBB permeability, confirming that activated NF-κB and increased ICAM-1 are essential factors involved in ischemia-induced BBB damage. Thus, our research provides a promising treatment strategy against BBB destruction after ischemic stroke, and TMEM16A may become a potential target for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Pin-Yi Liu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Zhi Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yi Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Xue-Lian Tang
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Shu Shu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Xin-Yu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yue Gu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| |
Collapse
|
48
|
Ali AA, Ahmed HI, Khaleel SA, Abu-Elfotuh K. Vinpocetine mitigates aluminum-induced cognitive impairment in socially isolated rats. Physiol Behav 2019; 208:112571. [PMID: 31175888 DOI: 10.1016/j.physbeh.2019.112571] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 05/11/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022]
Abstract
Several reports have highlighted the role of vinpocetine in Alzheimer's disease (AD). However, the role of vinpocetine in AD under social isolation conditions has not yet been elucidated. Henceforth, this study aimed to investigate the potential neuroprotective effect of vinpocetine in aluminum-induced AD model associated with social isolation. Social isolation increased the escape latency in Morris water maze (MWM) test, elevated the immobility score and decreased swimming score in forced swimming test (FST) in aluminum treated rats. However, vinpocetine enhanced acquisition in MWM test and exerted anti-depressive effect in FST. The histopathological examination showed marked deterioration in the cerebral cortex and hippocampus of AD isolated rats, while vinpocetine revealed overt improvement. In addition, the levels of amyloid-β protein (Aβ), phosphorylated-tau (Ser396), malondialdehyde (MDA), interleukin 1-beta (IL-1β), tumor necrosis alpha (TNFα), p- Glycogen synthase kinase-3β (p-GSK3β) (Tyr216), and β-secretase (BACE1) gene expression were increased in socially isolated aluminum treated rats, yet, vinpocetine treatment reversed these deteriorating effects. Hence, this study provides profound insights into the role of vinpocetine in AD particularly in the conditions of social isolation. The effects of vinpocetine might be attributed not only to its antioxidant and anti-inflammatory properties, but also to its suppressing effect on GSK3β activity and its downstream BACE1.
Collapse
Affiliation(s)
- Azza A Ali
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Hebatalla I Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt; Pharmacology and Toxicology Department, Faculty of Pharmacy, Heliopolis University for sustainable development, Cairo, Egypt
| | - Sahar A Khaleel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt; Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Karema Abu-Elfotuh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
49
|
Liu SP, Huang L, Flores J, Ding Y, Li P, Peng J, Zuo G, Zhang JH, Lu J, Tang JP. Secukinumab attenuates reactive astrogliosis via IL-17RA/(C/EBPβ)/SIRT1 pathway in a rat model of germinal matrix hemorrhage. CNS Neurosci Ther 2019; 25:1151-1161. [PMID: 31020769 PMCID: PMC6776744 DOI: 10.1111/cns.13144] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/28/2022] Open
Abstract
Aims Reactive astrogliosis plays a critical role in neurological deficits after germinal matrix hemorrhage (GMH). It has been reported that interleukin‐17A and IL‐17A receptor IL‐17RA/(C/EBPβ)/SIRT1 signaling pathway enhances reactive astrogliosis after brain injuries. We evaluated the effects of secukinumab on reactive astrogliosis in a rat pup model of GMH. Methods A total of 146 Sprague Dawley P7 rat pups were used. GMH was induced by intraparenchymal injection of collagenase. Secukinumab was administered intranasally 1 hour post‐GMH. C/EBPβ CRISPR or SIRT1 antagonist EX527 was administrated intracerebroventricularly (icv) 48 hours and 1 hour before GMH induction, respectively. Neurobehavior, Western blot, histology, and immunohistochemistry were used to assess treatment regiments in the short term and long term. Results The endogenous IL‐17A, IL‐17RA, C/EBPβ, and GFAP and proliferation marker CyclinD1 were increased, while SIRT1 expression was decreased after GMH. Secukinumab treatment improved neurological deficits, reduced ventriculomegaly, and increased cortical thickness. Additionally, treatment increased SIRT1 expression and lowered proliferation proteins PCNA and CyclinD1 as well as GFAP expression. C/EBPβ CRISPR activation plasmid and EX527 reversed the antireactive astrogliosis effects of secukinumab. Conclusion Secukinumab attenuated reactive astrogliosis and reduced neurological deficits after GMH, partly by regulating IL‐17RA/(C/EBPβ)/SIRT1 pathways. Secukinumab may provide a promising therapeutic strategy for GMH patients.
Collapse
Affiliation(s)
- Sheng-Peng Liu
- Department of Pediatrics, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China.,Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Lei Huang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jerry Flores
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Yan Ding
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Peng Li
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jun Peng
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Gang Zuo
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jun Lu
- Department of Pediatrics, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Ji-Ping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
50
|
Zhang X, Zhu XL, Ji BY, Cao X, Yu LJ, Zhang Y, Bao XY, Xu Y, Jin JL. LncRNA-1810034E14Rik reduces microglia activation in experimental ischemic stroke. J Neuroinflammation 2019; 16:75. [PMID: 30961627 PMCID: PMC6452518 DOI: 10.1186/s12974-019-1464-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/26/2019] [Indexed: 12/30/2022] Open
Abstract
Background Activation of microglial cells plays an important role in neuroinflammation after ischemic stroke. Inhibiting the activation of microglial cells has been suggested as a potential therapeutic approach in the treatment of ischemic stroke. Methods Oxygen-glucose deprivation in primary microglial cells and transient middle cerebral artery occlusion (MCAO) in C57BL/6 mice were used as the in vitro and in vivo ischemic stroke models. Microarray analysis was performed to investigate the overall impact of long non-coding RNAs (lncRNAs) on the inflammation status of microglial cells. RT-qPCR was used to evaluate the lncRNA levels and mRNA levels of cytokines and microglial cell markers. ELISA was taken to measure the level of cytokines. Immunofluorescence was used to observe the activation of microglial cells. Western blotting was performed to test the p65 phosphorylation. Results In this study, we showed that LncRNA-1810034E14Rik was significantly decreased in LPS-treated or oxygen-glucose deprivation-induced microglial cells. Overexpression of 1810034E14Rik decreased the infarct volume and alleviated brain damage in MCAO mice. 1810034E14Rik overexpression reduced the expression of inflammatory cytokines not only in ischemic stroke mice but also in oxygen-glucose deprivation-induced microglial cells. Moreover, 1810034E14Rik overexpression could suppress the activation of microglial cells and inhibit the phosphorylation of p65. Conclusions LncRNA-1810034E14Rik plays an anti-inflammatory role in ischemic stroke and regulates p65 phosphorylation, making it a potential target for stroke treatment. Electronic supplementary material The online version of this article (10.1186/s12974-019-1464-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Xiao-Lei Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Bi-Ying Ji
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Lin-Jie Yu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yan Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Xin-Yu Bao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China. .,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China. .,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, China. .,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, China.
| | - Jia-Li Jin
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China. .,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
| |
Collapse
|