1
|
Yang Y, Deng C, Aldali F, Huang Y, Luo H, Liu Y, Huang D, Cao X, Zhou Q, Xu J, Li Y, Chen H. Therapeutic Approaches and Potential Mechanisms of Small Extracellular Vesicles in Treating Vascular Dementia. Cells 2025; 14:409. [PMID: 40136659 PMCID: PMC11941715 DOI: 10.3390/cells14060409] [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: 01/12/2025] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Small extracellular vesicles (sEVs), including exosomes as a subtype, with a diameter typically less than 200 nm and originating from the endosomal system, are capable of transporting a diverse array of bioactive molecules, including proteins, nucleic acids, and lipids, thereby facilitating intercellular communication and modulating cellular functions. Vascular dementia (VaD) represents a form of cognitive impairment attributed to cerebrovascular disease, characterized by a complex and multifaceted pathophysiological mechanism. Currently, the therapeutic approach to VaD predominantly emphasizes symptom management, as no specific pharmacological treatment exists to cure the condition. Recent investigations have illuminated the significant role of sEVs in the pathogenesis of vascular dementia. This review seeks to provide a comprehensive analysis of the characteristics and functions of sEVs, with a particular focus on their involvement in vascular dementia and its underlying mechanisms. The objective is to advance the understanding of the interplays between sEVs and vascular dementia, thereby offering novel insights for future research and therapeutic strategies.
Collapse
Affiliation(s)
- Yujie Yang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Chunchu Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Fatima Aldali
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Yunjie Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Hongmei Luo
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Yizhou Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Danxia Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Xiaojian Cao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Qiuzhi Zhou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yajie Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
2
|
Rajab HA, Al-Kuraishy HM, Shokr MM, Al-Gareeb AI, Al-Harchan NA, Alruwaili M, Papadakis M, Alexiou A, Batiha GES. Statins for vascular dementia: A hype or hope. Neuroscience 2025; 567:45-55. [PMID: 39746645 DOI: 10.1016/j.neuroscience.2024.12.059] [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: 10/07/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Vascular dementia (VaD) is a second most common type of dementia subsequent to Alzheimer disease (AD). VaD is characterized by cognitive impairment and memory loss that may progress due to the development of cerebral amyloid angiopathy (CAA) a hallmark of AD. CAA triggers the progression of ischemic and hemorrhagic strokes with the subsequent the development of VaD and mixed dementia. Early diagnosis of patients with appropriate use of anti-inflammatory can prevent CAA-related inflammation and VaD development. Currently, there are no effective drugs in the management of VaD. Of note, cholesterol-lowering agent statins which are commonly used in patients with vascular diseases and dyslipidemia may affect the progression of VaD. Many previous studies highlighted the potential therapeutic efficacy of statins in treating VaD. Though, the underlying mechanisms of statins in prevention and treatment of VaD are not fully clarified. Consequently, this review aims to discuss the mechanistic role of statins in the management of VaD, and how statins may adversely affect the cognitive function in VaD patients.
Collapse
Affiliation(s)
- Hussein A Rajab
- Endocrinology Consultant, Medical School, Najran University, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Mustafa M Shokr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, Jabir Ibn Hayyan Medical University, Kufa, Iraq
| | - Nasser A Al-Harchan
- Department of Clinical Pharmacology, College of Dentistry, Al-Rasheed University, Baghdad, Iraq
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal, 42283, Germany.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia; Department of Research & Development, Funogen, Athens, Greece
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
3
|
Qiu Y, Cheng L, Xiong Y, Liu Z, Shen C, Wang L, Lu Y, Wei S, Zhang L, Yang SB, Zhang X. Advances in the Study of Necroptosis in Vascular Dementia: Focus on Blood-Brain Barrier and Neuroinflammation. CNS Neurosci Ther 2025; 31:e70224. [PMID: 39915907 PMCID: PMC11802338 DOI: 10.1111/cns.70224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/18/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Vascular dementia (VaD) includes a group of brain disorders that are characterized by cerebrovascular pathology.Neuroinflammation, disruption of the blood-brain barrier (BBB) permeability, white matter lesions, and neuronal loss are all significant pathological manifestations of VaD and play a key role in disease progression. Necroptosis, also known asprogrammed necrosis, is a mode of programmed cell death distinct from apoptosis and is closely associated with ischemic injury and neurodegenerative diseases. Recent studies have shown that necroptosis in VaD exacerbates BBB destruction, activates neuroinflammation, promotes neuronal loss, and severely affects VaD prognosis. RESULTS AND CONCLUSIONS In this review, we outline the significant roles of necroptosis and its molecular mechanisms in the pathological process of VaD, with a particular focus on the role of necroptosis in modulating neuroinflammation and exacerbating the disruption of BBB permeability in VaD, and elaborate on the molecular regulatory mechanisms and the centrally involved cells of necroptosis mediated by tumor necrosis factor-α in neuroinflammation in VaD. We also analyze the possibility and specific strategy that targeting necroptosis would help inhibit neuroinflammation and BBB destruction in VaD. With a focus on necroptosis, this study delved into its impact on the pathological changes and prognosis of VaD to provide new treatment ideas.
Collapse
Affiliation(s)
- Yuemin Qiu
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Lin Cheng
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Yinyi Xiong
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of RehabilitationAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Ziying Liu
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Chunxiao Shen
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Liangliang Wang
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Yujia Lu
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Shufei Wei
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Lushun Zhang
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Seung Bum Yang
- Department of Medical Non‐Commissioned OfficerWonkwang Health Science UniversityIksanRepublic of Korea
| | - Xiaorong Zhang
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| |
Collapse
|
4
|
Ye Q, Li X, Gao W, Gao J, Zheng L, Zhang M, Yang F, Li H. Role of Rho-associated kinases and their inhibitor fasudil in neurodegenerative diseases. Front Neurosci 2024; 18:1481983. [PMID: 39628659 PMCID: PMC11613983 DOI: 10.3389/fnins.2024.1481983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/01/2024] [Indexed: 12/06/2024] Open
Abstract
Neurodegenerative diseases (NDDs) are prevalent in the elderly. The pathogenesis of NDDs is complex, and currently, there is no cure available. With the increase in aging population, over 20 million people are affected by common NDDs alone (Alzheimer's disease and Parkinson's disease). Therefore, NDDs have profound negative impacts on patients, their families, and society, making them a major global health concern. Rho-associated kinases (ROCKs) belong to the serine/threonine protein kinases family, which modulate diverse cellular processes (e.g., apoptosis). ROCKs may elevate the risk of various NDDs (including Huntington's disease, Parkinson's disease, and Alzheimer's disease) by disrupting synaptic plasticity and promoting inflammatory responses. Therefore, ROCK inhibitors have been regarded as ideal therapies for NDDs in recent years. Fasudil, one of the classic ROCK inhibitor, is a potential drug for treating NDDs, as it repairs nerve damage and promotes axonal regeneration. Thus, the current review summarizes the relationship between ROCKs and NDDs and the mechanism by which fasudil inhibits ROCKs to provide new ideas for the treatment of NDDs.
Collapse
Affiliation(s)
- Qiuyan Ye
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xue Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Gao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
- Jiangsu College of Nursing, Huaian, China
| | - Jiayue Gao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Liping Zheng
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Miaomiao Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fengge Yang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Honglin Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
5
|
Lin L, Chen Y, He K, Metwally S, Jha R, Capuk O, Bhuiyan MIH, Singh G, Cao G, Yin Y, Sun D. Carotid artery vascular stenosis causes the blood-CSF barrier damage and neuroinflammation. J Neuroinflammation 2024; 21:220. [PMID: 39256783 PMCID: PMC11385148 DOI: 10.1186/s12974-024-03209-1] [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: 03/20/2024] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The choroid plexus (ChP) helps maintain the homeostasis of the brain by forming the blood-CSF barrier via tight junctions (TJ) at the choroid plexus epithelial cells, and subsequently preventing neuroinflammation by restricting immune cells infiltration into the central nervous system. However, whether chronic cerebral hypoperfusion causes ChP structural damage and blood-CSF barrier impairment remains understudied. METHODS The bilateral carotid stenosis (BCAS) model in adult male C57BL/6 J mice was used to induce cerebral hypoperfusion, a model for vascular contributions to cognitive impairment and dementia (VCID). BCAS-mediated changes of the blood-CSF barrier TJ proteins, apical secretory Na+-K+-Cl- cotransporter isoform 1 (NKCC1) protein and regulatory serine-threonine kinases SPAK, and brain infiltration of myeloid-derived immune cells were assessed. RESULTS BCAS triggered dynamic changes of TJ proteins (claudin 1, claudin 5) accompanied with stimulation of SPAK-NKCC1 complex and NF-κB in the ChP epithelial cells. These changes impacted the integrity of the blood-CSF barrier, as evidenced by ChP infiltration of macrophages/microglia, neutrophils and T cells. Importantly, pharmacological blockade of SPAK with its potent inhibitor ZT1a in BCAS mice attenuated brain immune cell infiltration and improved cognitive neurological function. CONCLUSIONS BCAS causes chronic ChP blood-CSF damage and immune cell infiltration. Our study sheds light on the SPAK-NKCC1 complex as a therapeutic target in neuroinflammation.
Collapse
Affiliation(s)
- Lin Lin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, 116027, Liaoning, China
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yang Chen
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kai He
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shamseldin Metwally
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Roshani Jha
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
| | - Okan Capuk
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Gazal Singh
- Biomedical Masters Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Research Service, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Yan Yin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, 116027, Liaoning, China.
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA.
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA.
- Research Service, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Lee DH, Lee EC, Park SW, Lee JY, Lee MR, Oh JS. Pathogenesis of Cerebral Small Vessel Disease: Role of the Glymphatic System Dysfunction. Int J Mol Sci 2024; 25:8752. [PMID: 39201439 PMCID: PMC11354389 DOI: 10.3390/ijms25168752] [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: 07/05/2024] [Revised: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Cerebral small vessel disease (CSVD) is a group of pathologies that affect the cerebral blood vessels. CSVD accounts for 25% of strokes and contributes to 45% of dementia. However, the pathogenesis of CSVD remains unclear, involving a variety of complex mechanisms. CSVD may result from dysfunction in the glymphatic system (GS). The GS contains aquaporin-4 (AQP-4), which is in the perivascular space, at the endfeet of the astrocyte. The GS contributes to the removal of waste products from the central nervous system, occupying perivascular spaces and regulating the exchange and movement of cerebrospinal fluid and interstitial fluid. The GS involves astrocytes and aquaporin channels, which are components of the blood-brain barrier, and problems with them may constitute the pathogenesis of CSVD. Vascular risk factors, including diabetes, dilate the perivascular space, disrupting the glymphatic system and the active regulation of AQP-4. CSVD exacerbation due to disorders of the GS is associated with multiple vasculopathies. Dysfunction of the glymphatic system and AQP-4 interferes with the functioning of the blood-brain barrier, which exacerbates CSVD. In a long-term follow-up of CSVD patients with microbleeds, lacunar infarcts, and white matter hyperintensity, several vascular risk factors, including hypertension, increased the risk of ischemic stroke. Dysfunction of the GS may be the cause of CSVD; however, the underlying treatment needs to be studied further.
Collapse
Affiliation(s)
- Dong-Hun Lee
- Industry-Academic Cooperation Foundation, The Catholic University of Korea, 222, Banpo-daro, Seocho-gu, Seoul 06591, Republic of Korea
| | - Eun Chae Lee
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang-Won Park
- Department of Neurosurgery, Uijeongbu St. Mary’s Hospital, College of Medicine, 271 Cheonbo-ro, Uijeongbu 11765, Republic of Korea
| | - Ji Young Lee
- Department of Neurosurgery, Uijeongbu St. Mary’s Hospital, College of Medicine, 271 Cheonbo-ro, Uijeongbu 11765, Republic of Korea
| | - Man Ryul Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Republic of Korea
| | - Jae Sang Oh
- Department of Neurosurgery, Uijeongbu St. Mary’s Hospital, College of Medicine, 271 Cheonbo-ro, Uijeongbu 11765, Republic of Korea
| |
Collapse
|
7
|
Wertman E. Essential New Complexity-Based Themes for Patient-Centered Diagnosis and Treatment of Dementia and Predementia in Older People: Multimorbidity and Multilevel Phenomenology. J Clin Med 2024; 13:4202. [PMID: 39064242 PMCID: PMC11277671 DOI: 10.3390/jcm13144202] [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: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Dementia is a highly prevalent condition with devastating clinical and socioeconomic sequela. It is expected to triple in prevalence by 2050. No treatment is currently known to be effective. Symptomatic late-onset dementia and predementia (SLODP) affects 95% of patients with the syndrome. In contrast to trials of pharmacological prevention, no treatment is suggested to remediate or cure these symptomatic patients. SLODP but not young onset dementia is intensely associated with multimorbidity (MUM), including brain-perturbating conditions (BPCs). Recent studies showed that MUM/BPCs have a major role in the pathogenesis of SLODP. Fortunately, most MUM/BPCs are medically treatable, and thus, their treatment may modify and improve SLODP, relieving suffering and reducing its clinical and socioeconomic threats. Regrettably, the complex system features of SLODP impede the diagnosis and treatment of the potentially remediable conditions (PRCs) associated with them, mainly due to failure of pattern recognition and a flawed diagnostic workup. We suggest incorporating two SLODP-specific conceptual themes into the diagnostic workup: MUM/BPC and multilevel phenomenological themes. By doing so, we were able to improve the diagnostic accuracy of SLODP components and optimize detecting and favorably treating PRCs. These revolutionary concepts and their implications for remediability and other parameters are discussed in the paper.
Collapse
Affiliation(s)
- Eli Wertman
- Department of Neurology, Hadassah University Hospital, The Hebrew University, Jerusalem 9190500, Israel;
- Section of Neuropsychology, Department of Psychology, The Hebrew University, Jerusalem 9190500, Israel
- Or’ad: Organization for Cognitive and Behavioral Changes in the Elderly, Jerusalem 9458118, Israel
- Merhav Neuropsychogeriatric Clinics, Nehalim 4995000, Israel
| |
Collapse
|
8
|
Dithmer S, Blasig IE, Fraser PA, Qin Z, Haseloff RF. The Basic Requirement of Tight Junction Proteins in Blood-Brain Barrier Function and Their Role in Pathologies. Int J Mol Sci 2024; 25:5601. [PMID: 38891789 PMCID: PMC11172262 DOI: 10.3390/ijms25115601] [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: 02/07/2024] [Revised: 03/10/2024] [Accepted: 03/28/2024] [Indexed: 06/21/2024] Open
Abstract
This review addresses the role of tight junction proteins at the blood-brain barrier (BBB). Their expression is described, and their role in physiological and pathological processes at the BBB is discussed. Based on this, new approaches are depicted for paracellular drug delivery and diagnostics in the treatment of cerebral diseases. Recent data provide convincing evidence that, in addition to its impairment in the course of diseases, the BBB could be involved in the aetiology of CNS disorders. Further progress will be expected based on new insights in tight junction protein structure and in their involvement in signalling pathways.
Collapse
Affiliation(s)
- Sophie Dithmer
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany (I.E.B.)
| | - Ingolf E. Blasig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany (I.E.B.)
| | | | - Zhihai Qin
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100049, China
| | - Reiner F. Haseloff
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany (I.E.B.)
| |
Collapse
|
9
|
He JB, Zhang H, Zheng HX, Jia JX, Zhang YC, Yan XS, Li XX, Wei KW, Mao J, Chen H, Li J, Wang H, Zhang M, Zhao ZY. Effects of schisandrin B on hypoxia-related cognitive function and protein expression in vascular dementia rats. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:421-427. [PMID: 38551405 DOI: 10.1080/15287394.2024.2334247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Vascular dementia (VD) a heterogenous group of brain disorders in which cognitive impairment is attributable to vascular risk factors and cerebrovascular disease. A common phenomenon in VD is a dysfunctional cerebral regulatory mechanism associated with insufficient cerebral blood flow, ischemia and hypoxia. Under hypoxic conditions oxygen supply to the brain results in neuronal death leading to neurodegenerative diseases including Alzheimer's (AD) and VD. In conditions of hypoxia and low oxygen perfusion, expression of hypoxia-inducible factor 1 alpha (HIF-1α) increases under conditions of low oxygen and low perfusion associated with upregulation of expression of hypoxia-upregulated mitochondrial movement regulator (HUMMR), which promotes anterograde mitochondrial transport by binding with trafficking protein kinesin 2 (TRAK2). Schisandrin B (Sch B) an active component derived from Chinese herb Wuweizi prevented β-amyloid protein induced morphological alterations and cell death using a SH-SY5Y neuronal cells considered an AD model. It was thus of interest to determine whether Sch B might also alleviate VD using a rat bilateral common carotid artery occlusion (BCAO) dementia model. The aim of this study was to examine the effects of Sch B in BCAO on cognitive functions such as Morris water maze test and underlying mechanisms involving expression of HIF-1α, TRAK2, and HUMMR levels. The results showed that Sch B improved learning and memory function of rats with VD and exerted a protective effect on the hippocampus by inhibition of protein expression of HIF-1α, TRAK2, and HUMMR factors. Evidence indicates that Sch B may be considered as an alternative in VD treatment.
Collapse
Affiliation(s)
- Jing-Bo He
- Department of Pharmacy, Baotou Medical College, Inner Mongolia, China
| | - He Zhang
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Hong-Xia Zheng
- Faculty of Foreign Languages, Baotou Teachers' College, Inner Mongolia, China
| | - Jian-Xin Jia
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Yi-Chi Zhang
- Class15, Senior two, Baotou No.9 High School, Inner Mongolia, China
| | - Xu-Sheng Yan
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Xiao-Xu Li
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Kai-Wen Wei
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Jun Mao
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Hong Chen
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Jing Li
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
- Department of Anesthesia, The Fourth Hospital of Inner Mongolia Autonomous Region, Inner Mongolia, China
| | - He Wang
- School of Health Sciences, University of Newcastle, Newcastle, Australia
| | - Ming Zhang
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Zhi-Ying Zhao
- Institute of Neuroscience, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| |
Collapse
|
10
|
Wang Y, Xie D, Ma S, Shao N, Zhang X, Wang X. Exploring the common mechanism of vascular dementia and inflammatory bowel disease: a bioinformatics-based study. Front Immunol 2024; 15:1347415. [PMID: 38736878 PMCID: PMC11084673 DOI: 10.3389/fimmu.2024.1347415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Objective Emerging evidence has shown that gut diseases can regulate the development and function of the immune, metabolic, and nervous systems through dynamic bidirectional communication on the brain-gut axis. However, the specific mechanism of intestinal diseases and vascular dementia (VD) remains unclear. We designed this study especially, to further clarify the connection between VD and inflammatory bowel disease (IBD) from bioinformatics analyses. Methods We downloaded Gene expression profiles for VD (GSE122063) and IBD (GSE47908, GSE179285) from the Gene Expression Omnibus (GEO) database. Then individual Gene Set Enrichment Analysis (GSEA) was used to confirm the connection between the two diseases respectively. The common differentially expressed genes (coDEGs) were identified, and the STRING database together with Cytoscape software were used to construct protein-protein interaction (PPI) network and core functional modules. We identified the hub genes by using the Cytohubba plugin. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were applied to identify pathways of coDEGs and hub genes. Subsequently, receiver operating characteristic (ROC) analysis was used to identify the diagnostic ability of these hub genes, and a training dataset was used to verify the expression levels of the hub genes. An alternative single-sample gene set enrichment (ssGSEA) algorithm was used to analyze immune cell infiltration between coDEGs and immune cells. Finally, the correlation between hub genes and immune cells was analyzed. Results We screened 167 coDEGs. The main articles of coDEGs enrichment analysis focused on immune function. 8 shared hub genes were identified, including PTPRC, ITGB2, CYBB, IL1B, TLR2, CASP1, IL10RA, and BTK. The functional categories of hub genes enrichment analysis were mainly involved in the regulation of immune function and neuroinflammatory response. Compared to the healthy controls, abnormal infiltration of immune cells was found in VD and IBD. We also found the correlation between 8 shared hub genes and immune cells. Conclusions This study suggests that IBD may be a new risk factor for VD. The 8 hub genes may predict the IBD complicated with VD. Immune-related coDEGS may be related to their association, which requires further research to prove.
Collapse
Affiliation(s)
- Yujiao Wang
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Daojun Xie
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Shijia Ma
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Nan Shao
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiaoyan Zhang
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xie Wang
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
11
|
Martos D, Lőrinczi B, Szatmári I, Vécsei L, Tanaka M. The Impact of C-3 Side Chain Modifications on Kynurenic Acid: A Behavioral Analysis of Its Analogs in the Motor Domain. Int J Mol Sci 2024; 25:3394. [PMID: 38542368 PMCID: PMC10970565 DOI: 10.3390/ijms25063394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 11/11/2024] Open
Abstract
The central nervous system (CNS) is the final frontier in drug delivery because of the blood-brain barrier (BBB), which poses significant barriers to the access of most drugs to their targets. Kynurenic acid (KYNA), a tryptophan (Trp) metabolite, plays an important role in behavioral functions, and abnormal KYNA levels have been observed in neuropsychiatric conditions. The current challenge lies in delivering KYNA to the CNS owing to its polar side chain. Recently, C-3 side chain-modified KYNA analogs have been shown to cross the BBB; however, it is unclear whether they retain the biological functions of the parent molecule. This study examined the impact of KYNA analogs, specifically, SZR-72, SZR-104, and the newly developed SZRG-21, on behavior. The analogs were administered intracerebroventricularly (i.c.v.), and their effects on the motor domain were compared with those of KYNA. Specifically, open-field (OF) and rotarod (RR) tests were employed to assess motor activity and skills. SZR-104 increased horizontal exploratory activity in the OF test at a dose of 0.04 μmol/4 μL, while SZR-72 decreased vertical activity at doses of 0.04 and 0.1 μmol/4 μL. In the RR test, however, neither KYNA nor its analogs showed any significant differences in motor skills at either dose. Side chain modification affects affective motor performance and exploratory behavior, as the results show for the first time. In this study, we showed that KYNA analogs alter emotional components such as motor-associated curiosity and emotions. Consequently, drug design necessitates the development of precise strategies to traverse the BBB while paying close attention to modifications in their effects on behavior.
Collapse
Affiliation(s)
- Diána Martos
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged, Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
| | - Bálint Lőrinczi
- Institute of Pharmaceutical Chemistry and HUN-REN–SZTE Stereochemistry Research Group, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (B.L.); (I.S.)
| | - István Szatmári
- Institute of Pharmaceutical Chemistry and HUN-REN–SZTE Stereochemistry Research Group, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (B.L.); (I.S.)
| | - László Vécsei
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged, Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged, Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
| |
Collapse
|
12
|
Tanaka M, Szabó Á, Körtési T, Szok D, Tajti J, Vécsei L. From CGRP to PACAP, VIP, and Beyond: Unraveling the Next Chapters in Migraine Treatment. Cells 2023; 12:2649. [PMID: 37998384 PMCID: PMC10670698 DOI: 10.3390/cells12222649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Migraine is a neurovascular disorder that can be debilitating for individuals and society. Current research focuses on finding effective analgesics and management strategies for migraines by targeting specific receptors and neuropeptides. Nonetheless, newly approved calcitonin gene-related peptide (CGRP) monoclonal antibodies (mAbs) have a 50% responder rate ranging from 27 to 71.0%, whereas CGRP receptor inhibitors have a 50% responder rate ranging from 56 to 71%. To address the need for novel therapeutic targets, researchers are exploring the potential of another secretin family peptide, pituitary adenylate cyclase-activating polypeptide (PACAP), as a ground-breaking treatment avenue for migraine. Preclinical models have revealed how PACAP affects the trigeminal system, which is implicated in headache disorders. Clinical studies have demonstrated the significance of PACAP in migraine pathophysiology; however, a few clinical trials remain inconclusive: the pituitary adenylate cyclase-activating peptide 1 receptor mAb, AMG 301 showed no benefit for migraine prevention, while the PACAP ligand mAb, Lu AG09222 significantly reduced the number of monthly migraine days over placebo in a phase 2 clinical trial. Meanwhile, another secretin family peptide vasoactive intestinal peptide (VIP) is gaining interest as a potential new target. In light of recent advances in PACAP research, we emphasize the potential of PACAP as a promising target for migraine treatment, highlighting the significance of exploring PACAP as a member of the antimigraine armamentarium, especially for patients who do not respond to or contraindicated to anti-CGRP therapies. By updating our knowledge of PACAP and its unique contribution to migraine pathophysiology, we can pave the way for reinforcing PACAP and other secretin peptides, including VIP, as a novel treatment option for migraines.
Collapse
Affiliation(s)
- Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
| | - Ágnes Szabó
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (Á.S.); (D.S.); (J.T.)
- Doctoral School of Clinical Medicine, University of Szeged, Korányi fasor 6, H-6720 Szeged, Hungary
| | - Tamás Körtési
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
- Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31, H-6726 Szeged, Hungary;
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| | - Délia Szok
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (Á.S.); (D.S.); (J.T.)
| | - János Tajti
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (Á.S.); (D.S.); (J.T.)
| | - László Vécsei
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (Á.S.); (D.S.); (J.T.)
| |
Collapse
|
13
|
Manukjan N, Majcher D, Leenders P, Caiment F, van Herwijnen M, Smeets HJ, Suidgeest E, van der Weerd L, Vanmierlo T, Jansen JFA, Backes WH, van Oostenbrugge RJ, Staals J, Fulton D, Ahmed Z, Blankesteijn WM, Foulquier S. Hypoxic oligodendrocyte precursor cell-derived VEGFA is associated with blood-brain barrier impairment. Acta Neuropathol Commun 2023; 11:128. [PMID: 37550790 PMCID: PMC10405482 DOI: 10.1186/s40478-023-01627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/23/2023] [Indexed: 08/09/2023] Open
Abstract
Cerebral small vessel disease is characterised by decreased cerebral blood flow and blood-brain barrier impairments which play a key role in the development of white matter lesions. We hypothesised that cerebral hypoperfusion causes local hypoxia, affecting oligodendrocyte precursor cell-endothelial cell signalling leading to blood-brain barrier dysfunction as an early mechanism for the development of white matter lesions. Bilateral carotid artery stenosis was used as a mouse model for cerebral hypoperfusion. Pimonidazole, a hypoxic cell marker, was injected prior to humane sacrifice at day 7. Myelin content, vascular density, blood-brain barrier leakages, and hypoxic cell density were quantified. Primary mouse oligodendrocyte precursor cells were exposed to hypoxia and RNA sequencing was performed. Vegfa gene expression and protein secretion was examined in an oligodendrocyte precursor cell line exposed to hypoxia. Additionally, human blood plasma VEGFA levels were measured and correlated to blood-brain barrier permeability in normal-appearing white matter and white matter lesions of cerebral small vessel disease patients and controls. Cerebral blood flow was reduced in the stenosis mice, with an increase in hypoxic cell number and blood-brain barrier leakages in the cortical areas but no changes in myelin content or vascular density. Vegfa upregulation was identified in hypoxic oligodendrocyte precursor cells, which was mediated via Hif1α and Epas1. In humans, VEGFA plasma levels were increased in patients versus controls. VEGFA plasma levels were associated with increased blood-brain barrier permeability in normal appearing white matter of patients. Cerebral hypoperfusion mediates hypoxia induced VEGFA expression in oligodendrocyte precursor cells through Hif1α/Epas1 signalling. VEGFA could in turn increase BBB permeability. In humans, increased VEGFA plasma levels in cerebral small vessel disease patients were associated with increased blood-brain barrier permeability in the normal appearing white matter. Our results support a role of VEGFA expression in cerebral hypoperfusion as seen in cerebral small vessel disease.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Daria Majcher
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Peter Leenders
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Marcel van Herwijnen
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Hubert J. Smeets
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Ernst Suidgeest
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
| | - Louise van der Weerd
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, The Netherlands
| | - Tim Vanmierlo
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, European Graduate School of Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Jacobus F. A. Jansen
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Walter H. Backes
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Robert J. van Oostenbrugge
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Julie Staals
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
- Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
14
|
Choi SO, Choi JG, Yun JY. A Study of Brain Function Characteristics of Service Members at High Risk for Accidents in the Military. Brain Sci 2023; 13:1157. [PMID: 37626513 PMCID: PMC10452066 DOI: 10.3390/brainsci13081157] [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: 06/03/2023] [Revised: 07/23/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Military accidents are often associated with stress and depressive psychological conditions among soldiers, and they often fail to adapt to military life. Therefore, this study analyzes whether there are differences in EEG and pulse wave indices between general soldiers and three groups of soldiers who have not adapted to military life and are at risk of accidents. Data collection was carried out using a questionnaire and a device that can measure EEG and pulse waves, and data analysis was performed using SPSS. The results showed that the concentration level and brain activity indices were higher in the general soldiers and the soldiers in the first stage of accident risk. The body stress index was higher for each stage of accident risk, and the physical vitality index was higher for general soldiers. Therefore, it can be seen that soldiers who have not adapted to military life and are at risk of accidents have somewhat lower concentration and brain activity than general soldiers, and have symptoms of stress and lethargy. The results of this study will contribute to reducing human accidents through EEG and pulse wave measurements not only in the military but also in occupations with a high risk of accidents such as construction.
Collapse
Affiliation(s)
| | | | - Jong-Yong Yun
- Department of Protection and Safety Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| |
Collapse
|
15
|
Killick R, Elliott C, Ribe E, Broadstock M, Ballard C, Aarsland D, Williams G. Neurodegenerative Disease Associated Pathways in the Brains of Triple Transgenic Alzheimer's Model Mice Are Reversed Following Two Weeks of Peripheral Administration of Fasudil. Int J Mol Sci 2023; 24:11219. [PMID: 37446396 PMCID: PMC10342807 DOI: 10.3390/ijms241311219] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
The pan Rho-associated coiled-coil-containing protein kinase (ROCK) inhibitor fasudil acts as a vasodilator and has been used as a medication for post-cerebral stroke for the past 29 years in Japan and China. More recently, based on the involvement of ROCK inhibition in synaptic function, neuronal survival, and processes associated with neuroinflammation, it has been suggested that the drug may be repurposed for neurodegenerative diseases. Indeed, fasudil has demonstrated preclinical efficacy in many neurodegenerative disease models. To facilitate an understanding of the wider biological processes at play due to ROCK inhibition in the context of neurodegeneration, we performed a global gene expression analysis on the brains of Alzheimer's disease model mice treated with fasudil via peripheral IP injection. We then performed a comparative analysis of the fasudil-driven transcriptional profile with profiles generated from a meta-analysis of multiple neurodegenerative diseases. Our results show that fasudil tends to drive gene expression in a reverse sense to that seen in brains with post-mortem neurodegenerative disease. The results are most striking in terms of pathway enrichment analysis, where pathways perturbed in Alzheimer's and Parkinson's diseases are overwhelmingly driven in the opposite direction by fasudil treatment. Thus, our results bolster the repurposing potential of fasudil by demonstrating an anti-neurodegenerative phenotype in a disease context and highlight the potential of in vivo transcriptional profiling of drug activity.
Collapse
Affiliation(s)
- Richard Killick
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK; (R.K.); (E.R.); (D.A.)
- College of Medicine and Health, University of Exeter, Exeter EX1 2UL, UK;
| | - Christina Elliott
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Elena Ribe
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK; (R.K.); (E.R.); (D.A.)
| | - Martin Broadstock
- Wolfson CARD, King’s College London, London Bridge, London SE1 1UL, UK;
| | - Clive Ballard
- College of Medicine and Health, University of Exeter, Exeter EX1 2UL, UK;
| | - Dag Aarsland
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK; (R.K.); (E.R.); (D.A.)
| | - Gareth Williams
- Wolfson CARD, King’s College London, London Bridge, London SE1 1UL, UK;
| |
Collapse
|
16
|
Taha M, Eldemerdash OM, Elshaffei IM, Yousef EM, Soliman AS, Senousy MA. Apigenin Attenuates Hippocampal Microglial Activation and Restores Cognitive Function in Methotrexate-Treated Rats: Targeting the miR-15a/ROCK-1/ERK1/2 Pathway. Mol Neurobiol 2023; 60:3770-3787. [PMID: 36943623 DOI: 10.1007/s12035-023-03299-7] [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: 11/14/2022] [Accepted: 02/22/2023] [Indexed: 03/23/2023]
Abstract
Microglial activation underpins the methotrexate (MTX)-induced neurotoxicity; however, the precise mechanism remains unclear. This study appraised the potential impact of apigenin (Api), a neuroprotective flavonoid, in MTX-induced neurotoxicity in rats in terms of microglial activation through targeting the miR-15a/Rho-associated protein kinase-1 (ROCK-1)/extracellular signal-regulated kinase 1/2 (ERK1/2) pathway. Male Sprague Dawley rats were randomly divided into 4 groups: Normal control (saline i.p. daily and i.v. on days 8 and 15); Api control (20 mg/kg, p.o.) daily for 30 days; MTX-alone (75 mg/kg, i.v.) on days 8 and 15, then four i.p. injections of leucovorin (LCV): 6 mg/kg after 18 h, then three doses (3 mg/kg) every 8 h post-MTX; and Api co-treated (20 mg/kg/day, p.o.) throughout the model for 30 days, with administration of MTX and LCV as in group 3. MTX administration elevated hippocampal ionized calcium-binding adaptor protein-1 (Iba-1) immunostaining, indicating microglial activation. This was accompanied by neuroinflammation, oxidative stress, and enhanced apoptosis manifested by elevated hippocampal interleukin-1β, malondialdehyde, and caspase-3, and decreased reduced glutathione levels. Concurrently, abated miR-15a expression, overexpression of its target ROCK-1, diminished downstream ERK1/2 and cAMP response element-binding protein (CREB) phosphorylation, and decreased hippocampal brain-derived neurotrophic factor (BDNF) levels were observed. Api mitigated the MTX-induced neurotoxicity by reversing the biochemical, histopathological, and behavioral derangements tested by novel object recognition and Morris water maze tests. Conclusively, Api lessens MTX-induced neuroinflammation, oxidative stress, and apoptosis and boosts cognitive function through inhibiting microglial activation via modulating the miR-15a/ROCK-1/ERK1/2/CREB/BDNF pathway. Graphical abstract showing the effects of methotrexate and apigenin co-treatment in MTX-induced neurotoxicity model. On the left, methotrexate (MTX) administration to rats resulted in hippocampal miR-15a downregulation, which triggered an enhanced expression of its target ROCK-1, consequently inhibiting the downstream ERK1/2/CREB/BDNF pathway, instigating a state of microglial activation, neuroinflammation, oxidative stress, and apoptosis. On the other hand, apigenin (Api) co-treatment restored miR-15a, inhibited ROCK-1 expression, and activated the ERK1/2/CREB/BDNF pathway, leading to diminished hippocampal microglial activation, neuroinflammation, and apoptosis, and restoration of the redox balance, along with improvement in memory and cognitive function of the MTX-treated rats.
Collapse
Affiliation(s)
- Mohamed Taha
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Kasr El Ainy st., Cairo, 11562, Egypt.
| | - Omar Mohsen Eldemerdash
- Department of Biochemistry, Faculty of Pharmacy, Misr International University (MIU), KM 28 Cairo, Ismailia Road, Cairo, 44971, Egypt
| | - Ismail Mohamed Elshaffei
- Department of Biochemistry, Faculty of Pharmacy, Misr International University (MIU), KM 28 Cairo, Ismailia Road, Cairo, 44971, Egypt
| | - Einas Mohamed Yousef
- Department of Histology and Cell Biology, Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
| | - Ayman S Soliman
- Medical Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Mahmoud Ahmed Senousy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Kasr El Ainy st., Cairo, 11562, Egypt
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt
| |
Collapse
|
17
|
Zhu SJ, Li X, Wei YW, Luo YF, Tang GH, Tang ZS. Acupoint catgut embedding improves learning and memory impairment in vascular dementia rats. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:108. [PMID: 36819587 PMCID: PMC9929827 DOI: 10.21037/atm-22-6402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023]
Abstract
Background Vascular dementia (VD) is a disease that affects brain function through cerebrovascular disease. Due to its complex pathogenesis, there is no effective drug treatment for VD. The present study aimed to evaluate the role of acupoint catgut embedding in the treatment of rats with VD and its possible molecular mechanism. Methods A modified 4 vessel occlusion (4-VO) method was used to establish a VD model rat, and spatial learning and memory ability was assessed using the Morris water maze (MWM) test. The protein expression levels were detected by Western blot. Hematoxylin and eosin (HE) staining was used for histological analysis and enzyme-linked immunosorbent assay (ELISA) was applied for analysis of serum inflammatory factors. Results We successfully constructed VD model rats with spatial learning and memory impairment, hippocampus injury, and high inflammatory response. Treatment of VD rats with acupoint catgut embedding significantly reduced escape latency and increased the time in the target quadrant and platform crossing times. VD-mediated hippocampal tissue damage and inflammatory reaction [down-regulating interleukin-1β (IL-1β), interleukin-6 (IL-6)] were significantly alleviated by acupoint catgut embedding treatment. In addition, further mechanism exploration found that acupoint catgut embedding treatment could improve the activity of the toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88)/nuclear factor-κB (NF-κB) signaling pathway. In summary, acupoint catgut embedding treatment improved spatial learning and memory loss, alleviated pathological damage of the hippocampus, and inhibited inflammation response in VD rats, which was probably related to the inhibition of the TLR4/MyD88/NF-κB signaling pathway. Conclusions Acupoint catgut embedding may warrant further study as an adjuvant therapy for the treatment of VD.
Collapse
Affiliation(s)
- Shi-Jie Zhu
- Anatomy Teaching and Research Office of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xia Li
- Intelligent Elderly Care Teaching and Research Office of Guiyang Health Care Vocational University, Guiyang, China
| | - Yu-Wei Wei
- First Department of Gerontology, The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine (Hanan Branch), Harbin, China
| | - Ya-Fei Luo
- Anatomy Teaching and Research Office of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Gui-Hua Tang
- Department of Neurology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhong-Sheng Tang
- Anatomy Teaching and Research Office of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
18
|
Wu F, Li Y, Liu W, Xiao R, Yao B, Gao M, Xu D, Wang J. Comparative Investigation of Raw and Processed Radix Polygoni Multiflori on the Treatment of Vascular Dementia by Liquid Chromatograph-Mass Spectrometry Based Metabolomic Approach. Metabolites 2022; 12:metabo12121297. [PMID: 36557335 PMCID: PMC9785642 DOI: 10.3390/metabo12121297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/07/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Radix Polygoni Multiflori (PM) is a well-known nootropic used in traditional Chinese medicine (TCM). Considering the efficacy and application discrepancy between raw (RPM) and processed PM (PPM), the similarities and differences between them in the treatment of vascular dementia (VaD) is intriguing. In this study, a VaD rat model was constructed by 2-vessel occlusion (2-VO). During 28 days of treatment, plasma was collected on days 7, 14, 21, and 28 after the start of dosing and the metabolic profile was analyzed by HPLC-MS/MS-based metabolomics. The Morris Water Maze Test, hematoxylin-eosin and Nissl staining, and biochemical analysis were used to assess cognitive function, pathogenic alterations and oxidative stress, respectively. RPM and PPM effectivelyreducedthe 2VO-induced cognitive impairment and mitigated histological alterations in hippocampus tissue. The 2-VO model significantly elevated MDA level and decreased SOD activity and GSH level, indicating severe oxidative stress, which could also be attenuated by RPM and PPM treatment. RPM outperformed PPM in decreasing MDA levels while PPM outperformed RPM in increasing GSH levels. Differential metabolites were subjected to Metabolite Set Enrichment Analysis (MSEA) and genes corresponding to proteins having interactions with metabolites were further annotated with Gene Ontology (GO). Both RPM and PPM ameliorated VaD-relevant vitamin B6 metabolism, pentose phosphate pathways, and taurine and hypotaurine metabolism. In addition, the metabolism of cysteine and methionine was regulated only by RPM, and riboflavin metabolism was modulated only by PPM. The results suggested that raw and processed PM had comparable efficacy in the treatment of VaD but also with some mechanistic differenece.
Collapse
|
19
|
Tanaka M, Szabó Á, Vécsei L. Integrating Armchair, Bench, and Bedside Research for Behavioral Neurology and Neuropsychiatry: Editorial. Biomedicines 2022; 10:2999. [PMID: 36551755 PMCID: PMC9775182 DOI: 10.3390/biomedicines10122999] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
"To learning much inclined, who went to see the Elephant (though all of them were blind) that each by observation might satisfy the mind" [...].
Collapse
Affiliation(s)
- Masaru Tanaka
- ELKH-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Ágnes Szabó
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Korányi Fasor 6, H-6720 Szeged, Hungary
| | - László Vécsei
- ELKH-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
20
|
Tanaka M, Vécsei L. Editorial of Special Issue ‘Dissecting Neurological and Neuropsychiatric Diseases: Neurodegeneration and Neuroprotection’. Int J Mol Sci 2022; 23:ijms23136991. [PMID: 35805990 PMCID: PMC9266548 DOI: 10.3390/ijms23136991] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023] Open
Affiliation(s)
- Masaru Tanaka
- ELKH-SZTE Neuroscience Research Group, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Semmelweis u. 6, H-6725 Szeged, Hungary
| | - László Vécsei
- ELKH-SZTE Neuroscience Research Group, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Semmelweis u. 6, H-6725 Szeged, Hungary
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
21
|
Lee DH, Lee JY, Hong DY, Lee EC, Park SW, Jo YN, Park YJ, Cho JY, Cho YJ, Chae SH, Lee MR, Oh JS. ROCK and PDE-5 Inhibitors for the Treatment of Dementia: Literature Review and Meta-Analysis. Biomedicines 2022; 10:biomedicines10061348. [PMID: 35740369 PMCID: PMC9219677 DOI: 10.3390/biomedicines10061348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/02/2022] [Accepted: 06/05/2022] [Indexed: 12/14/2022] Open
Abstract
Dementia is a disease in which memory, thought, and behavior-related disorders progress gradually due to brain damage caused by injury or disease. It is mainly caused by Alzheimer’s disease or vascular dementia and several other risk factors, including genetic factors. It is difficult to treat as its incidence continues to increase worldwide. Many studies have been performed concerning the treatment of this condition. Rho-associated kinase (ROCK) and phosphodiesterase-5 (PDE-5) are attracting attention as pharmacological treatments to improve the symptoms. This review discusses how ROCK and PDE-5 affect Alzheimer’s disease, vascular restructuring, and exacerbation of neuroinflammation, and how their inhibition helps improve cognitive function. In addition, the results of the animal behavior analysis experiments utilizing the Morris water maze were compared through meta-analysis to analyze the effects of ROCK inhibitors and PDE-5 inhibitors on cognitive function. According to the selection criteria, 997 publications on ROCK and 1772 publications on PDE-5 were screened, and conclusions were drawn through meta-analysis. Both inhibitors showed good improvement in cognitive function tests, and what is expected of the synergy effect of the two drugs was confirmed in this review.
Collapse
Affiliation(s)
- Dong-Hun Lee
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Ji Young Lee
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
| | - Dong-Yong Hong
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Eun Chae Lee
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Sang-Won Park
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Yu Na Jo
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Yu Jin Park
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Jae Young Cho
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Yoo Jin Cho
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Su Hyun Chae
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Man Ryul Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
- Correspondence: (M.R.L.); (J.S.O.)
| | - Jae Sang Oh
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
- Correspondence: (M.R.L.); (J.S.O.)
| |
Collapse
|
22
|
Martos D, Tuka B, Tanaka M, Vécsei L, Telegdy G. Memory Enhancement with Kynurenic Acid and Its Mechanisms in Neurotransmission. Biomedicines 2022; 10:biomedicines10040849. [PMID: 35453599 PMCID: PMC9027307 DOI: 10.3390/biomedicines10040849] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 03/28/2022] [Accepted: 04/02/2022] [Indexed: 02/04/2023] Open
Abstract
Kynurenic acid (KYNA) is an endogenous tryptophan (Trp) metabolite known to possess neuroprotective property. KYNA plays critical roles in nociception, neurodegeneration, and neuroinflammation. A lower level of KYNA is observed in patients with neurodegenerative diseases such as Alzheimer’s and Parkinson’s diseases or psychiatric disorders such as depression and autism spectrum disorders, whereas a higher level of KYNA is associated with the pathogenesis of schizophrenia. Little is known about the optimal concentration for neuroprotection and the threshold for neurotoxicity. In this study the effects of KYNA on memory functions were investigated by passive avoidance test in mice. Six different doses of KYNA were administered intracerebroventricularly to previously trained CFLP mice and they were observed for 24 h. High doses of KYNA (i.e., 20–40 μg/2 μL) significantly decreased the avoidance latency, whereas a low dose of KYNA (0.5 μg/2 μL) significantly elevated it compared with controls, suggesting that the low dose of KYNA enhanced memory function. Furthermore, six different receptor blockers were applied to reveal the mechanisms underlying the memory enhancement induced by KYNA. The series of tests revealed the possible involvement of the serotonergic, dopaminergic, α and β adrenergic, and opiate systems in the nootropic effect. This study confirmed that a low dose of KYNA improved a memory component of cognitive domain, which was mediated by, at least in part, four systems of neurotransmission in an animal model of learning and memory.
Collapse
Affiliation(s)
- Diána Martos
- MTA-SZTE Neuroscience Research Group, Hungarian Academy of Sciences, University of Szeged (MTA-SZTE), Semmelweis u. 6, H-6725 Szeged, Hungary; (D.M.); (B.T.); (M.T.)
| | - Bernadett Tuka
- MTA-SZTE Neuroscience Research Group, Hungarian Academy of Sciences, University of Szeged (MTA-SZTE), Semmelweis u. 6, H-6725 Szeged, Hungary; (D.M.); (B.T.); (M.T.)
| | - Masaru Tanaka
- MTA-SZTE Neuroscience Research Group, Hungarian Academy of Sciences, University of Szeged (MTA-SZTE), Semmelweis u. 6, H-6725 Szeged, Hungary; (D.M.); (B.T.); (M.T.)
| | - László Vécsei
- MTA-SZTE Neuroscience Research Group, Hungarian Academy of Sciences, University of Szeged (MTA-SZTE), Semmelweis u. 6, H-6725 Szeged, Hungary; (D.M.); (B.T.); (M.T.)
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-342-361
| | - Gyula Telegdy
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 5, H-6725 Szeged, Hungary;
| |
Collapse
|