1
|
Bogadi S, Bhaskaran M, Ravichandran V, Nesamony J, Chelliah S, Kuppusamy G, Prakash GM, Karri VVSR, Mallick S, Farahim F, Ali T, Babu DR, Subramaniyan V. Functionalized Nanoparticles: A Promising Approach for Effective Management of Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04917-2. [PMID: 40234291 DOI: 10.1007/s12035-025-04917-2] [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: 12/01/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
The severe neurodegenerative disease known as Alzheimer's disease (AD) is typified by a progressive loss of memory and cognitive function. The prevalence of AD is rising due to an aging global population, calling for novel treatment strategies. A potential treatment option for AD that shows promise is the use of functionalized nanoparticles (NPs). Recent developments in the synthesis, design, and use of functionalized NPs in AD therapy are examined in this review. An outline of the pathophysiological mechanisms underlying AD is given in the first section, focusing on the roles played by tau protein aggregates and amyloid-beta plaques in the development of the illness. We then explore the many approaches used to functionalize NPs, such as surface alterations and bioconjugation methods, which enable accurate drug administration, targeted delivery, and enhanced biocompatibility. The review also emphasizes the therapeutic potential of functionalized NPs, highlighting their capacity to improve neuroprotection, lower amyloid-beta aggregation, and improve blood-brain barrier penetration. The potential of NPs as a tool for disease modification and symptom relief is highlighted by recent pre-clinical and clinical research. Concerns about toxicity and safety are also covered, underscoring the significance of thorough testing and the field's future directions. Functionalized NPs have great promise as a multimodal strategy to treat AD, offering patients hope for better quality of life, early diagnosis, and efficient disease treatment. This study highlights the growing role of nanotechnology in the search for novel and potent therapies for AD.
Collapse
Affiliation(s)
- Subhasri Bogadi
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, OotyNilgiris, Tamil Nadu, India
| | - Mahendran Bhaskaran
- College of Pharmacy and Pharmaceutical Sciences, University of Toledo HSC, 3000 Arlington Avenue, Toledo, OH, 43614, USA.
| | - Vishnuvardh Ravichandran
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
| | - Jerry Nesamony
- College of Pharmacy and Pharmaceutical Sciences, University of Toledo HSC, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Selvam Chelliah
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX- 77004, USA
| | - Gowthamarajan Kuppusamy
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, OotyNilgiris, Tamil Nadu, India
| | - Gowrav Mysore Prakash
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, OotyNilgiris, Tamil Nadu, India
| | | | - Samir Mallick
- Tennessee State University, Chemistry department, 3500 John A Merritt Blvd, Nashville, TN, 37209, USA
| | - Farha Farahim
- Department of Nursing, King Khalid University, Abha, 61413, Kingdom of Saudi Arabia
| | - Talat Ali
- Department of Basic Medical Sciences, King Khalid University, Abha, 61413, Kingdom of Saudi Arabia
| | | | - Vetriselvan Subramaniyan
- Department of Pharmacology, Jeffrey Cheah School of Medicine and Health Sciences MONASH University, Subang Jaya, Malaysia
| |
Collapse
|
2
|
Yılmaz BÖ, Şencan R. The effect of fibrinogen levels on three-month neurological recovery in acute ischemic stroke patients. Sci Rep 2025; 15:12644. [PMID: 40221477 PMCID: PMC11993669 DOI: 10.1038/s41598-025-91611-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 02/21/2025] [Indexed: 04/14/2025] Open
Abstract
The present study aims to examine the relationship between post-stroke fibrinogen levels and long-term (three-month) prognosis in patients who had their first ischemic stroke and to determine the effect of fibrinogen levels on morbidity and mortality. A prospective study was carried out on 100 patients presenting with their first ischemic stroke at the Neurology Clinic of Gaziantep City Hospital. Stroke severity was examined utilizing the National Institutes of Health Stroke Scale (NIHSS) and the modified Rankin Scale (mRS) at initial admission and at third month post-stroke. Serum fibrinogen levels were recorded from routine blood tests, and the relationship between fibrinogen levels and clinical outcomes was analyzed statistically. Significant positive correlations were observed between fibrinogen levels and stroke severity and outcomes. Fibrinogen levels were associated with NIHSS admission scores (r = 0.371, p < 0.001) and mRS admission scores (r = 0.439, p < 0.001). At third month, fibrinogen levels were significantly related with NIHSS (r = 0.544, p < 0.001) and mRS (r = 0.538, p < 0.001) scores. Regression analysis revealed that a one-unit increase in fibrinogen levels resulted in a 0.018-unit increase in NIHSS scores and a 0.008-unit increase in mRS scores (p < 0.001). Elevated fibrinogen levels during the acute phase are related with poorer neurological outcomes and increased morbidity and mortality within the three-month follow-up period in patients with first-time ischemic stroke. These resultsindicate that fibrinogen could be considered a prognostic biomarker for stroke management and rehabilitation planning.
Collapse
Affiliation(s)
- Buket Özkara Yılmaz
- Gaziantep City Hospital Department of Neurology, İbn-i Sina District, 27470, Sahinbey/Gaziantep, Turkey.
| | - Ramazan Şencan
- Gaziantep City Hospital Department of Neurology, İbn-i Sina District, 27470, Sahinbey/Gaziantep, Turkey
| |
Collapse
|
3
|
Al-Kuraishy HM, Sulaiman GM, Mohammed HA, Mohammed SG, Al-Gareeb AI, Albuhadily AK, Dawood RA, Al Ali A, Abu-Alghayth MH. Amyloid-β and heart failure in Alzheimer's disease: the new vistas. Front Med (Lausanne) 2025; 12:1494101. [PMID: 39967593 PMCID: PMC11832649 DOI: 10.3389/fmed.2025.1494101] [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: 09/10/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and represents 75% of all dementia types. AD neuropathology is due to the progressive deposition of extracellular amyloid-beta (Aβ) peptide and intracellular hyperphosphorylated tau protein. The accumulated Aβ forms amyloid plaques, while the hyperphosphorylated tau protein forms neurofibrillary tangles (NFTs). Both amyloid plaques and NFTs are hallmarks of AD neuropathology. The fundamental mechanism involved in the pathogenesis of AD is still elusive, although Aβ is the more conceivable theory. Aβ-induced neurodegeneration and associated neuroinflammation, oxidative stress, endoplasmic reticulum stress (ER), and mitochondrial dysfunction contribute to the development of cognitive impairment and dementia. Of note, Aβ is not only originated from the brain but also produced peripherally and, via the blood-brain barrier (BBB), can accumulate in the brain and result in the development of AD. It has been shown that cardiometabolic conditions such as obesity, type 2 diabetes (T2D), and heart failure (HF) are regarded as possible risk factors for the development of AD and other types of dementia, such as vascular dementia. HF-induced chronic cerebral hypoperfusion, oxidative stress, and inflammation can induce the development and progression of AD. Interestingly, AD is regarded as a systemic disease that causes systemic inflammation and oxidative stress, which in turn affects peripheral organs, including the heart. Aβ through deranged BBB can be transported into the systemic circulation from the brain and accumulated in the heart, leading to the development of HF. These findings suggest a close relationship between AD and HF. However, the exact mechanism of AD-induced HF is not fully elucidated. Therefore, this review aims to discuss the link between AD and the risk of HF regarding the potential role of Aβ in the pathogenesis of HF.
Collapse
Affiliation(s)
- Hayder M. Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | | | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim, Saudi Arabia
| | - Sohaib G. Mohammed
- Department of Pathological Analysis, College of Applied Science, Samarra University, Saladin, Iraq
| | | | - Ali K. Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Retaj A. Dawood
- Department of Biology, College of Science, Al-Mustaqbal University, Hilla, Iraq
| | - Amer Al Ali
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Mohammed H. Abu-Alghayth
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| |
Collapse
|
4
|
Luo S, Wang Y, Hisatsune T. P2Y1 receptor in Alzheimer's disease. Neural Regen Res 2025; 20:440-453. [PMID: 38819047 PMCID: PMC11317937 DOI: 10.4103/nrr.nrr-d-23-02103] [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: 12/29/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is the most frequent form of dementia characterized by the deposition of amyloid-beta plaques and neurofibrillary tangles consisting of hyperphosphorylated tau. Targeting amyloid-beta plaques has been a primary direction for developing Alzheimer's disease treatments in the last decades. However, existing drugs targeting amyloid-beta plaques have not fully yielded the expected results in the clinic, necessitating the exploration of alternative therapeutic strategies. Increasing evidence unravels that astrocyte morphology and function alter in the brain of Alzheimer's disease patients, with dysregulated astrocytic purinergic receptors, particularly the P2Y1 receptor, all of which constitute the pathophysiology of Alzheimer's disease. These receptors are not only crucial for maintaining normal astrocyte function but are also highly implicated in neuroinflammation in Alzheimer's disease. This review delves into recent insights into the association between P2Y1 receptor and Alzheimer's disease to underscore the potential neuroprotective role of P2Y1 receptor in Alzheimer's disease by mitigating neuroinflammation, thus offering promising avenues for developing drugs for Alzheimer's disease and potentially contributing to the development of more effective treatments.
Collapse
Affiliation(s)
- Shan Luo
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Yifei Wang
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Tatsuhiro Hisatsune
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| |
Collapse
|
5
|
Power L, Shuhmaher R, Houtz P, Chen J, Rudolph S, Yuen J, Machour M, Levy E, Wu L, Levenberg S, Whalen M, Chen Y, Kaplan DL. 3D Neurovascular Unit Tissue Model to Assess Responses to Traumatic Brain Injury. J Biomed Mater Res A 2025; 113:e37816. [PMID: 39440483 DOI: 10.1002/jbm.a.37816] [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: 07/09/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
The neurovascular unit (NVU) is a critical interface in the central nervous system that links vascular interactions with glial and neural tissue. Disruption of the NVU has been linked to the onset and progression of neurodegenerative diseases. Despite its significance the NVU remains challenging to study in a physiologically relevant manner. Here, a 3D cell triculture model of the NVU is developed that incorporates human primary brain microvascular endothelial cells, astrocytes, and pericytes into a tissue system that can be sustained in vitro for several weeks. This tissue model helps recapitulate the complexity of the NVU and can be used to interrogate the mechanisms of disease and cell-cell interactions. The NVU tissue model displays elevated cell death and inflammatory responses following mechanical damage, to emulate traumatic brain injury (TBI) under controlled laboratory conditions, including lactate dehydrogenase (LDH) release, elevated inflammatory markers TNF-α and monocyte chemoattractant cytokines MCP-2 and MCP-3 and reduced expression of the tight junction marker ZO-1. This 3D tissue model serves as a tool for deciphering mechanisms of TBIs and immune responses associated with the NVU.
Collapse
Affiliation(s)
- Liam Power
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Rita Shuhmaher
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Philip Houtz
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Jinpeng Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - John Yuen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Majd Machour
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Emily Levy
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Limin Wu
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Michael Whalen
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
6
|
Patel S, Govindarajan V, Chakravarty S, Dubey N. From blood to brain: Exploring the role of fibrinogen in the pathophysiology of depression and other neurological disorders. Int Immunopharmacol 2024; 143:113326. [PMID: 39388892 DOI: 10.1016/j.intimp.2024.113326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
Recent findings indicate that fibrinogen, a protein involved in blood clotting, plays a significant role in neuroinflammation and mood disorders. Elevated fibrinogen levels are consistently observed in individuals with depression, potentially contributing to microglial activation. This could impair fibrinolysis and contribute to a pro-inflammatory environment in the brain. This neuroinflammatory response can impair neuroplasticity, a key process for learning, memory, and mood regulation. Fibrinogen may also indirectly influence neurotransmitters like serotonin, which play a vital role in mood regulation. Furthermore, fibrinogen's interaction with astrocytes may trigger a cascade of events leading to demyelination, a process where the protective sheath around nerve fibers deteriorates. This can disrupt communication within the nervous system and contribute to depression symptoms. Intriguingly, targeting fibrinogen or related pathways holds promise for therapeutic interventions. For instance, modulating PAI-1 (Plasminogen activator inhibitor-1) activity or inhibiting fibrinogen's interaction with brain cells could be potential strategies. This review explores the multifaceted relationship between fibrinogen and neurological disorders with a focus on depression highlighting its potential as a therapeutic target. Further research is necessary to fully elucidate the mechanisms underlying this association and develop effective therapeutic strategies targeting the fibrinolytic system for mood disorders.
Collapse
Affiliation(s)
- Shashikant Patel
- Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, India
| | - Venkatesh Govindarajan
- Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad 500046, Telangana, India
| | - Sumana Chakravarty
- Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, India.
| | - Neelima Dubey
- Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad 500046, Telangana, India.
| |
Collapse
|
7
|
Zheng H, Zheng H, Wei L, Xue Z, Xu B, Hu M, Yu J, Xie R, Zhang L, Zheng Z, Xie J, Zheng C, Huang C, Lin J, Li P. Risk stratification models incorporating oxidative stress factors to predict survival and recurrence in patients with gastric cancer after radical gastrectomy: A real-world multicenter study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108658. [PMID: 39244978 DOI: 10.1016/j.ejso.2024.108658] [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: 05/13/2024] [Revised: 08/13/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Oxidative stress significantly influences the development and progression of gastric cancer (GC). It remains unreported whether incorporating oxidative stress factors into nomograms can improve the predictive accuracy for survival and recurrence risk in GC patients. METHODS 3498 GC patients who underwent radical gastrectomy between 2009 and 2017 were enrolled and randomly divided into training cohort (TC) and internal validation cohort (IVC). Cox regression analysis model was used to evaluate six preoperative oxidative stress indicators to formulate the Systemic oxidative stress Score (SOSS). Two nomograms based on SOSS was constructed by multivariate Cox regression and validated using 322 patients from another two hospitals. RESULTS A total of 3820 patients were included. The SOSS, composed of three preoperative indicators-fibrinogen, albumin, and cholesterol-was an independent prognostic factor for both overall survival (OS) and disease-free survival (DFS). The two nomograms based on SOSS showed a significantly higher AUC than the pTNM stage (OS: 0.830 vs. 0.778, DFS: 0.824 vs. 0.775, all P < 0.001) and were validated in the IVC and EVC (all P < 0.001). The local recurrence rate, peritoneal recurrence rate, distant recurrence rate and multiple recurrence rate in high-risk group were significantly higher than those in low-risk group (P < 0.05). CONCLUSIONS The two novel nomograms based on SOSS which was a combination score of three preoperative blood indicators, demonstrated outstanding predictive abilities for both survival and recurrence in GC patients with different risk groups, which may potentially improve survival through perioperatively active intervention strategies and individualized postoperatively close surveillance.
Collapse
Affiliation(s)
- Honghong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Hualong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Linghua Wei
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Zhen Xue
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Binbin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Minggao Hu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Department of General Surgery, The PLA Navy Anqing Hospital, Anqing, 246000, China
| | - Junhua Yu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, 324000, China
| | - Rongzhen Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, 321000, China
| | - Lingkang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Zhiwei Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Jianwei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - ChangMing Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China.
| | - Jianxian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China.
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China.
| |
Collapse
|
8
|
Yue Q, Leng X, Xie N, Zhang Z, Yang D, Hoi MPM. Endothelial Dysfunctions in Blood-Brain Barrier Breakdown in Alzheimer's Disease: From Mechanisms to Potential Therapies. CNS Neurosci Ther 2024; 30:e70079. [PMID: 39548663 PMCID: PMC11567945 DOI: 10.1111/cns.70079] [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: 07/17/2024] [Revised: 09/13/2024] [Accepted: 09/28/2024] [Indexed: 11/18/2024] Open
Abstract
Recent research has shown the presence of blood-brain barrier (BBB) breakdown in Alzheimer's disease (AD). BBB is a dynamic interface consisting of a continuous monolayer of brain endothelial cells (BECs) enveloped by pericytes and astrocytes. The restricted permeability of BBB strictly controls the exchange of substances between blood and brain parenchyma, which is crucial for brain homeostasis by excluding blood-derived detrimental factors and pumping out brain-derived toxic molecules. BBB breakdown in AD is featured as a series of BEC pathologies such as increased paracellular permeability, abnormal levels and functions of transporters, and inflammatory or oxidative profile, which may disturb the substance transportation across BBB, thereafter induce CNS disorders such as hypometabolism, Aβ accumulation, and neuroinflammation, eventually aggravate cognitive decline. Therefore, it seems important to protect BEC properties for BBB maintenance and neuroprotection. In this review, we thoroughly summarized the pathological alterations of BEC properties reported in AD patients and numerous AD models, including paracellular permeability, influx and efflux transporters, and inflammatory and oxidative profiles, and probably associated underlying mechanisms. Then we reviewed current therapeutic agents that are effective in ameliorating a series of BEC pathologies, and ultimately protecting BBB integrity and cognitive functions. Regarding the current drug development for AD proceeds extremely hard, this review aims to discuss the therapeutic potentials of targeting BEC pathologies and BBB maintenance for AD treatment, therefore expecting to shed a light on the future AD drug development by targeting BEC pathologies and BBB protection.
Collapse
Affiliation(s)
- Qian Yue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacao SARChina
- Department of Pharmaceutical Sciences, Faculty of Health SciencesUniversity of MacauMacao SARChina
- Department of CardiologyThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
- The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital)HeyuanGuangdongChina
| | - Xinyue Leng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacao SARChina
- Department of Pharmaceutical Sciences, Faculty of Health SciencesUniversity of MacauMacao SARChina
| | - Ningqing Xie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio‐Cerebrovascular Diseases, and Institute of New Drug ResearchJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs ResearchJinan University College of PharmacyGuangzhouChina
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)Jinan University College of PharmacyGuangzhouChina
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio‐Cerebrovascular Diseases, and Institute of New Drug ResearchJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs ResearchJinan University College of PharmacyGuangzhouChina
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)Jinan University College of PharmacyGuangzhouChina
| | - Deguang Yang
- Department of CardiologyThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
- The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital)HeyuanGuangdongChina
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)Jinan University College of PharmacyGuangzhouChina
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacao SARChina
- Department of Pharmaceutical Sciences, Faculty of Health SciencesUniversity of MacauMacao SARChina
| |
Collapse
|
9
|
Osborne OM, Daftari M, Naranjo O, Johar AN, Brooks S, Colbert BM, Torices S, Lewis E, Sendaydiego J, Drexler G, Bashti M, Margetts AV, Tuesta LM, Mason C, Bilbao D, Vontell R, Griswold AJ, Dykxhoorn DM, Toborek M. Post-stroke hippocampal neurogenesis is impaired by microvascular dysfunction and PI3K signaling in cerebral amyloid angiopathy. Cell Rep 2024; 43:114848. [PMID: 39392753 PMCID: PMC11562893 DOI: 10.1016/j.celrep.2024.114848] [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: 04/30/2024] [Revised: 07/05/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024] Open
Abstract
Ischemic stroke and cerebral amyloid angiopathy (CAA) pose significant challenges in an aging population, particularly in post-stroke recovery. Using the 5xFAD mouse model, we explore the relationship between CAA, ischemic stroke, and tissue recovery. We hypothesize that amyloid-beta accumulation worsens stroke outcomes by inducing blood-brain barrier (BBB) dysfunction, leading to impaired neurogenesis. Our findings show that CAA exacerbates stroke outcomes, with mice exhibiting constricted BBB microvessels, reduced cerebral blood flow, and impaired tissue recovery. Transcriptional analysis shows that endothelial cells and neural progenitor cells (NPCs) in the hippocampus exhibit differential gene expression in response to CAA and stroke, specifically targeting the phosphatidylinositol 3-kinase (PI3K) pathway. In vitro experiments with human NPCs validate these findings, showing that disruption of the CXCL12-PIK3C2A-CREB3L2 axis impairs neurogenesis. Notably, PI3K pathway activation restores neurogenesis, highlighting a potential therapeutic approach. These results suggest that CAA combined with stroke induces microvascular dysfunction and aberrant neurogenesis through this specific pathway.
Collapse
Affiliation(s)
- Olivia M Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Manav Daftari
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Adarsh N Johar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Samantha Brooks
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Brett M Colbert
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elizabeth Lewis
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jet Sendaydiego
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gillian Drexler
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Malek Bashti
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander V Margetts
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Luis M Tuesta
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurology and Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christian Mason
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Regina Vontell
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA; Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Anthony J Griswold
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Derek M Dykxhoorn
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
10
|
Wang J, Huo X, Zhou H, Liu H, Li X, Lu N, Sun X. Identification of Autophagy-Related Candidate Genes in the Early Diagnosis of Alzheimer's Disease and Exploration of Potential Molecular Mechanisms. Mol Neurobiol 2024; 61:6584-6598. [PMID: 38329682 DOI: 10.1007/s12035-024-04011-z] [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: 09/21/2023] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
This study aimed to identify autophagy-related candidate genes for the early diagnosis of Alzheimer's disease (AD) and elucidate their potential molecular mechanisms. Differentially expressed genes (DEGs) and phenotype-associated significant module genes were obtained using the "limma" package and weighted gene co-expression network analysis (WGCNA) based on hippocampal tissue datasets from AD patients and control samples. The intersection between the list of autophagy-related genes (ATGs), DEGs, and module genes was further investigated to obtain AD-autophagy-related differential expression genes (ATDEGs). Subsequently, the least absolute shrinkage and selection operator (LASSO) algorithm was utilized to identify hub genes, and a second intersection was performed with important module genes from the protein-protein interaction (PPI) network to obtain co-hub genes. Finally, a diagnostic model was constructed by receiver operating characteristic (ROC) analysis to determine the candidate genes with high diagnostic efficacy in the external validation set. Moreover, immune infiltration analysis was performed on AD patient brain tissues and explore the correlation between candidate genes and immune cells. We further analyzed the expression level of candidate genes in the SH-SY5Y cells with Aβ25-35 (25 µM). Among the 17 identified AD-ATDEGs, ATP6V1E1 stood out with area under the curve (AUC) values of 0.869, 0.817, and 0.714 in the external validation set, underscoring its high diagnostic efficacy in both hippocampal and peripheral blood contexts for AD patients. Meanwhile, ATP6V1E1 expression was positively correlated with effector memory CD4 + T cells, while negatively correlated with natural killer T cells and activated CD4 + T cells. Results from quantitative PCR (qPCR) and immunofluorescence assays indicated a reduction in ATP6V1E1 expression, aligning with our database analysis findings. In summary, ATP6V1E1 as a candidate gene provides a new perspective for the early identification and pathogenesis of AD.
Collapse
Affiliation(s)
- Jian Wang
- The Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, China.
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China.
- Hunan Guangxiu Medical Imaging Diagnosis Center, Changsha, China.
| | - Xinhua Huo
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Huiqin Zhou
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Huasheng Liu
- Department of Radiology, Central South University, The Third Xiangya Hospital, Changsha, China
| | - Xiaofeng Li
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Na Lu
- Reproductive and Genetic Hospital of CITIC Xiangya, Changsha, China
| | - Xuan Sun
- The Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
11
|
Ma LY, Song JH, Gao PY, Ou YN, Fu Y, Huang LY, Wang ZT, Zhang DD, Cui RP, Mi YC, Tan L. Amyloid pathology mediates the associations between plasma fibrinogen and cognition in non-demented adults. J Neurochem 2024; 168:2532-2542. [PMID: 38533619 DOI: 10.1111/jnc.16105] [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] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
Though previous studies revealed the potential associations of elevated levels of plasma fibrinogen with dementia, there is still limited understanding regarding the influence of Alzheimer's disease (AD) biomarkers on these associations. We sought to investigate the interrelationships among fibrinogen, cerebrospinal fluid (CSF) AD biomarkers, and cognition in non-demented adults. We included 1996 non-demented adults from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study and 337 from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. The associations of fibrinogen with AD biomarkers and cognition were explored using multiple linear regression models. The mediation analyses with 10 000 bootstrapped iterations were conducted to explore the mediating effects of AD biomarkers on cognition. In addition, interaction analyses and subgroup analyses were conducted to assess the influence of covariates on the relationships between fibrinogen and AD biomarkers. Participants exhibiting low Aβ42 were designated as A+, while those demonstrating high phosphorylated tau (P-tau) and total tau (Tau) were labeled as T+ and N+, respectively. Individuals with normal measures of Aβ42 and P-tau were categorized as the A-T- group, and those with abnormal levels of both Aβ42 and P-tau were grouped under A+T+. Fibrinogen was higher in the A+ subgroup compared to that in the A- subgroup (p = 0.026). Fibrinogen was higher in the A+T+ subgroup compared to that in the A-T- subgroup (p = 0.011). Higher fibrinogen was associated with worse cognition and Aβ pathology (all p < 0.05). Additionally, the associations between fibrinogen and cognition were partially mediated by Aβ pathology (mediation proportion range 8%-28%). Interaction analyses and subgroup analyses showed that age and ApoE ε4 affect the relationships between fibrinogen and Aβ pathology. Fibrinogen was associated with both cognition and Aβ pathology. Aβ pathology may be a critical mediator for impacts of fibrinogen on cognition.
Collapse
Affiliation(s)
- Li-Yun Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jing-Hui Song
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Dan-Dan Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Rui-Ping Cui
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yin-Chu Mi
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Hossen F, Sun GY, Lee JC. Oligomeric Tau-induced oxidative damage and functional alterations in cerebral endothelial cells: Role of RhoA/ROCK signaling pathway. Free Radic Biol Med 2024; 221:261-272. [PMID: 38815773 PMCID: PMC11184584 DOI: 10.1016/j.freeradbiomed.2024.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/22/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Despite of yet unknown mechanism, microvascular deposition of oligomeric Tau (oTau) has been implicated in alteration of the Blood-Brain Barrier (BBB) function in Alzheimer's disease (AD) brains. In this study, we employed an in vitro BBB model using primary mouse cerebral endothelial cells (CECs) to investigate the mechanism underlying the effects of oTau on BBB function. We found that exposing CECs to oTau induced oxidative stress through NADPH oxidase, increased oxidative damage to proteins, decreased proteasome activity, and expressions of tight junction (TJ) proteins including occludin, zonula occludens-1 (ZO-1) and claudin-5. These effects were suppressed by the pretreatment with Fasudil, a RhoA/ROCK signaling inhibitor. Consistent with the biochemical alterations, we found that exposing the basolateral side of CECs to oTau in the BBB model disrupted the integrity of the BBB, as indicated by an increase in FITC-dextran transport across the model, and a decrease in trans endothelial electrical resistance (TEER). oTau also increased the transmigration of peripheral blood mononuclear cells (PBMCs) in the BBB model. These functional alterations in the BBB induced by oTau were also suppressed by Fasudil. Taken together, our findings suggest that targeting the RhoA/ROCK pathway can be a potential therapeutic strategy to maintain BBB function in AD.
Collapse
Affiliation(s)
- Faruk Hossen
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60607, USA
| | - Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia, MO, 65211, USA
| | - James C Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
13
|
Li F, Gallego J, Tirko NN, Greaser J, Bashe D, Patel R, Shaker E, Van Valkenburg GE, Alsubhi AS, Wellman S, Singh V, Padilla CG, Gheres KW, Broussard JI, Bagwell R, Mulvihill M, Kozai TDY. Low-intensity pulsed ultrasound stimulation (LIPUS) modulates microglial activation following intracortical microelectrode implantation. Nat Commun 2024; 15:5512. [PMID: 38951525 PMCID: PMC11217463 DOI: 10.1038/s41467-024-49709-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/13/2024] [Indexed: 07/03/2024] Open
Abstract
Microglia are important players in surveillance and repair of the brain. Implanting an electrode into the cortex activates microglia, produces an inflammatory cascade, triggers the foreign body response, and opens the blood-brain barrier. These changes can impede intracortical brain-computer interfaces performance. Using two-photon imaging of implanted microelectrodes, we test the hypothesis that low-intensity pulsed ultrasound stimulation can reduce microglia-mediated neuroinflammation following the implantation of microelectrodes. In the first week of treatment, we found that low-intensity pulsed ultrasound stimulation increased microglia migration speed by 128%, enhanced microglia expansion area by 109%, and a reduction in microglial activation by 17%, indicating improved tissue healing and surveillance. Microglial coverage of the microelectrode was reduced by 50% and astrocytic scarring by 36% resulting in an increase in recording performance at chronic time. The data indicate that low-intensity pulsed ultrasound stimulation helps reduce the foreign body response around chronic intracortical microelectrodes.
Collapse
Affiliation(s)
- Fan Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
- Computational Modeling and Simulation PhD Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jazlyn Gallego
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Natasha N Tirko
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | | | - Derek Bashe
- Washington University in St. Louis, St. Louis, MO, USA
| | - Rudra Patel
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric Shaker
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | - Vanshika Singh
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Camila Garcia Padilla
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | | | | | | | | | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA.
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Sun Y, Jiang M, Long X, Miao Y, Du H, Zhang T, Ma X, Zhang Y, Meng H. Transcriptomic Analysis of Lipid Metabolism Genes in Alzheimer's Disease: Highlighting Pathological Outcomes and Compartmentalized Immune Status. J Mol Neurosci 2024; 74:55. [PMID: 38776015 DOI: 10.1007/s12031-024-02225-3] [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: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 07/20/2024]
Abstract
The dysregulation of lipid metabolism has been strongly associated with Alzheimer's disease (AD) and has intricate connections with various aspects of disease progression, such as amyloidogenesis, bioenergetic deficit, oxidative stress, neuroinflammation, and myelin degeneration. Here, a comprehensive bioinformatic assessment was conducted on lipid metabolism genes in the brains and peripheral blood of AD-derived transcriptome datasets, characterizing the correlation between differentially expressed genes (DEGs) of lipid metabolism and disease pathologies, as well as immune cell preferences. Through the application of weighted gene co-expression network analysis (WGCNA), modules eigengenes related to lipid metabolism were pinpointed, and the examination of their molecular functions within biological processes, molecular pathways, and their associations with pathological phenotypes and molecular networks has been characterized. Analysis of biological networks indicates notable discrepancies in the expression patterns of the DEGs between neuronal and immune cells, as well as variations in cell type enrichments within both brain tissue and peripheral blood. Additionally, drugs targeting the DEGs from central and peripheral and a diagnostic model for hub genes from the blood were retrieved and assessed, some of which were shown to be useful for therapeutic and diagnostic. These results revealed the distinctive pattern of transcriptionally abnormal lipid metabolism in central, peripheral, and immune cell activation, providing valuable insight into lipid metabolism for diagnosing and guiding more effective treatment for AD.
Collapse
Affiliation(s)
- Yue Sun
- Department of Neurology, Shanghai East Hospital of Tongji University School of Medicine, Shanghai, 20092, People's Republic of China
| | - Mengni Jiang
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Xiang Long
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Yongzhen Miao
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Huanhuan Du
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Ting Zhang
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Xuejun Ma
- School of Mathematical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yue Zhang
- Department of Neurology, Shanghai East Hospital of Tongji University School of Medicine, Shanghai, 20092, People's Republic of China.
| | - Hongrui Meng
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China.
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.
| |
Collapse
|
15
|
Hasan I, Guo B, Zhang J, Chang C. Advances in Antioxidant Nanomedicines for Imaging and Therapy of Alzheimer's Disease. Antioxid Redox Signal 2024; 40:863-888. [PMID: 36070437 DOI: 10.1089/ars.2022.0107] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Reactive oxygen species (ROS) are crucial signaling molecules in the regulation of numerous physiological activities including the formation and function of the central nervous system (CNS). So far, many functional antioxidant nanomedicines with ROS scavenging capability to reduce oxidative stress in Alzheimer's disease (AD) have been developed for both imaging and therapy of AD. Recent Advances: This review focuses on the most recent advances in antioxidant nanomedicines such as ROS-scavenging nanoparticles (NPs), NPs with intrinsic antioxidant activity, and drug-loaded antioxidant NPs for AD theranostics. In addition to antioxidant nanomedicines, the emerging phototherapy treatment paradigms and the promising preclinic drug carriers, such as exosomes and liposomes, are also introduced. Critical Issues: In general, excessive generation of ROS can cause lipid peroxidation, oxidative DNA, as well as protein damage, aggravating pathogenic alterations, accumulation of amyloid-beta plaques and neurofibrillary tangles in the brain. These negative factors further cause cell death, which is the beginning of AD. Future Directions: We anticipate that this review will help researchers in the area of preclinical research and clinical translation of antioxidant nanomedicines for AD imaging and therapy.
Collapse
Affiliation(s)
- Ikram Hasan
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
| | - Bing Guo
- School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen, China
| | - Jian Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Chunqi Chang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
16
|
Hu Q, Zhang R, Dong X, Yang D, Yu W, Du Q. Huperzine A ameliorates neurological deficits after spontaneous subarachnoid hemorrhage through endothelial cell pyroptosis inhibition. Acta Biochim Biophys Sin (Shanghai) 2024; 56:645-656. [PMID: 38529553 DOI: 10.3724/abbs.2024037] [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] [Indexed: 03/27/2024] Open
Abstract
Spontaneous subarachnoid hemorrhage (SAH) is a kind of hemorrhagic stroke which causes neurological deficits in survivors. Huperzine A has a neuroprotective effect, but its role in SAH is unclear. Therefore, we explore the effect of Huperzine A on neurological deficits induced by SAH and the related mechanism. In this study, Evans blue assay, TUNEL staining, immunofluorescence, western blot analysis, and ELISA are conducted. We find that Huperzine A can improve neurological deficits and inhibit the apoptosis of nerve cells in SAH rats. Huperzine A treatment can improve the upregulation of brain water content, damage of blood-brain barrier, fibrinogen and matrix metalloprotein 9 expressions and the downregulation of ZO-1 and occludin expressions induced by SAH. Huperzine A inhibit the expressions of proteins involved in pyroptosis in endothelial cells in SAH rats. The increase in MDA content and decrease in SOD activity in SAH rats can be partly reversed by Huperzine A. The ROS inducer H 2O 2 can induce pyroptosis and inhibit the expressions of ZO-1 and occludin in endothelial cells, which can be blocked by Huperzine A. In addition, the increase in the entry of p65 into the nucleus in endothelial cells can be partly reversed by Huperzine A. Huperzine A may delay the damage of blood-brain barrier in SAH rats by inhibiting oxidative stress-mediated pyroptosis and tight junction protein expression downregulation through the NF-κB pathway. Overall, Huperzine A may have clinical value for treating SAH.
Collapse
Affiliation(s)
- Qiang Hu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Rong Zhang
- Medical Examination Center, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
| | - Xiaoqiao Dong
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Dingbo Yang
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Wenhua Yu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| |
Collapse
|
17
|
Bejarano L, Kauzlaric A, Lamprou E, Lourenco J, Fournier N, Ballabio M, Colotti R, Maas R, Galland S, Massara M, Soukup K, Lilja J, Brouland JP, Hottinger AF, Daniel RT, Hegi ME, Joyce JA. Interrogation of endothelial and mural cells in brain metastasis reveals key immune-regulatory mechanisms. Cancer Cell 2024; 42:378-395.e10. [PMID: 38242126 DOI: 10.1016/j.ccell.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/11/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024]
Abstract
Brain metastasis (BrM) is a common malignancy, predominantly originating from lung, melanoma, and breast cancers. The vasculature is a key component of the BrM tumor microenvironment with critical roles in regulating metastatic seeding and progression. However, the heterogeneity of the major BrM vascular components, namely endothelial and mural cells, is still poorly understood. We perform single-cell and bulk RNA-sequencing of sorted vascular cell types and detect multiple subtypes enriched specifically in BrM compared to non-tumor brain, including previously unrecognized immune regulatory subtypes. We integrate the human data with mouse models, creating a platform to interrogate vascular targets for the treatment of BrM. We find that the CD276 immune checkpoint molecule is significantly upregulated in the BrM vasculature, and anti-CD276 blocking antibodies prolonged survival in preclinical trials. This study provides important insights into the complex interactions between the vasculature, immune cells, and cancer cells, with translational relevance for designing therapeutic interventions.
Collapse
Affiliation(s)
- Leire Bejarano
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Annamaria Kauzlaric
- Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Eleni Lamprou
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Joao Lourenco
- Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nadine Fournier
- Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Michelle Ballabio
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland
| | - Roberto Colotti
- In Vivo Imaging Facility (IVIF), University of Lausanne, Lausanne, Switzerland
| | - Roeltje Maas
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Sabine Galland
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Matteo Massara
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Klara Soukup
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland
| | - Johanna Lilja
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland
| | - Jean-Philippe Brouland
- Department of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Andreas F Hottinger
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Roy T Daniel
- Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Monika E Hegi
- Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Neuroscience Research Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| |
Collapse
|
18
|
Li F, Gallego J, Tirko NN, Greaser J, Bashe D, Patel R, Shaker E, Van Valkenburg GE, Alsubhi AS, Wellman S, Singh V, Padill CG, Gheres KW, Bagwell R, Mulvihill M, Kozai TDY. Low-intensity pulsed ultrasound stimulation (LIPUS) modulates microglial activation following intracortical microelectrode implantation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570162. [PMID: 38105969 PMCID: PMC10723293 DOI: 10.1101/2023.12.05.570162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Microglia are important players in surveillance and repair of the brain. Their activation mediates neuroinflammation caused by intracortical microelectrode implantation, which impedes the application of intracortical brain-computer interfaces (BCIs). While low-intensity pulsed ultrasound stimulation (LIPUS) can attenuate microglial activation, its potential to modulate the microglia-mediated neuroinflammation and enhance the bio-integration of microelectrodes remains insufficiently explored. We found that LIPUS increased microglia migration speed from 0.59±0.04 to 1.35±0.07 µm/hr on day 1 and enhanced microglia expansion area from 44.50±6.86 to 93.15±8.77 µm 2 /min on day 7, indicating improved tissue healing and surveillance. Furthermore, LIPUS reduced microglial activation by 17% on day 6, vessel-associated microglia ratio from 70.67±6.15 to 40.43±3.87% on day 7, and vessel diameter by 20% on day 28. Additionally, microglial coverage of the microelectrode was reduced by 50% in week 1, indicating better tissue-microelectrode integration. These data reveal that LIPUS helps resolve neuroinflammation around chronic intracortical microelectrodes.
Collapse
|
19
|
Alruwaili M, Al-kuraishy HM, Alexiou A, Papadakis M, ALRashdi BM, Elhussieny O, Saad HM, Batiha GES. Pathogenic Role of Fibrinogen in the Neuropathology of Multiple Sclerosis: A Tale of Sorrows and Fears. Neurochem Res 2023; 48:3255-3269. [PMID: 37442896 PMCID: PMC10514123 DOI: 10.1007/s11064-023-03981-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating neurodegenerative disease of the central nervous system (CNS) due to injury of the myelin sheath by immune cells. The clotting factor fibrinogen is involved in the pathogenesis of MS by triggering microglia and the progress of neuroinflammation. Fibrinogen level is correlated with MS severity; consequently, inhibition of the fibrinogen cascade may reduce MS neuropathology. Thus, this review aimed to clarify the potential role of fibrinogen in the pathogenesis of MS and how targeting of fibrinogen affects MS neuropathology. Accumulation of fibrinogen in the CNS may occur independently or due to disruption of blood-brain barrier (BBB) integrity in MS. Fibrinogen acts as transduction and increases microglia activation which induces the progression of inflammation, oxidative stress, and neuronal injury. Besides, brain fibrinogen impairs the remyelination process by inhibiting the differentiation of oligodendrocyte precursor cells. These findings proposed that fibrinogen is associated with MS neuropathology through interruption of BBB integrity, induction of neuroinflammation, and demyelination with inhibition of the remyelination process by suppressing oligodendrocytes. Therefore, targeting of fibrinogen and/or CD11b/CD18 receptors by metformin and statins might decrease MS neuropathology. In conclusion, inhibiting the expression of CD11b/CD18 receptors by metformin and statins may decrease the pro-inflammatory effect of fibrinogen on microglia which is involved in the progression of MS.
Collapse
Affiliation(s)
- Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770 Australia
- AFNP Med, 1030 Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283 Wuppertal, Germany
| | - Barakat M. ALRashdi
- Biology Department, College of Science, Jouf University, Sakaka, 41412 Saudi Arabia
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744 Egypt
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744 Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Egypt
| |
Collapse
|
20
|
Liu J, Lai S, Wu P, Wang J, Wang J, Wang J, Zhang Y. Systematic oxidative stress indices predicts prognosis in patients with urothelial carcinoma of the upper urinary tract after radical nephroureterectomy. Eur J Med Res 2023; 28:469. [PMID: 37898799 PMCID: PMC10612206 DOI: 10.1186/s40001-023-01295-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/17/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Oxidative stress plays an important role in the occurrence and development of malignancy. However, the relationship between oxidative stress and upper urinary tract urothelial carcinoma (UTUC) prognosis remains elusive. This study aimed to evaluate the prognostic value of systematic oxidative stress indices as a predictor of patient outcomes in UTUC after radical nephroureterectomy. METHODS Clinical data for 483 patients with UTUC who underwent radical nephroureterectomy were analyzed. Patients were categorized according to an optimal value of systematic oxidative stress indices (SOSIs), including fibrinogen (Fib), gamma-glutamyl transpeptidase (γ-GGT), creatinine (CRE), lactate dehydrogenase (LDH) and albumin (ALB). Kaplan-Meier analyses were used to investigate associations of SOSIs with overall survival (OS) and progression-free survival (PFS). Moreover, associations between SOSIs and OS and PFS were assessed with univariate and multivariate analyses. RESULTS High values of Fib, γ-GGT, CRE, and LDH, and low values of ALB were associated with reduced OS. SOSIs status correlated with age, tumor site, surgical approach, hydronephrosis, tumor size, T stage, and lymph node status. The Kaplan-Meier survival analysis showed a significant discriminatory ability for death and progression risks in the two groups based on SOSIs. Multivariate Cox proportional hazards models showed that SOSIs were an independent prognostic indicator for OS (p = 0.007) and PFS (p = 0.021). SOSIs and clinical variables were selected to establish a nomogram for OS. The 1-, 3-, and 5-year AUC values were 0.77, 0.78, and 0.81, respectively. Calibration curves of the nomogram showed high consistencies between the predicted and observed survival probability. Decision curve analysis curves showed that the nomogram could well predict the 1-year, 3-year, and 5-year OS. CONCLUSIONS SOSIs are an independent unfavorable predictor of OS and PFS in patients diagnosed with UTUC undergoing RNU. Therefore, incorporating SOSIs into currently available clinical parameters may improve clinical decision-making.
Collapse
Affiliation(s)
- Jianyong Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Hospital Continence Center, Beijing, People's Republic of China
| | - Shicong Lai
- Department of Urology, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Pengjie Wu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Hospital Continence Center, Beijing, People's Republic of China
| | - Jiawen Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Beijing Hospital Continence Center, Beijing, People's Republic of China
| | - Jianye Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China.
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
- Beijing Hospital Continence Center, Beijing, People's Republic of China.
| | - Jianlong Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China.
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
- Beijing Hospital Continence Center, Beijing, People's Republic of China.
| | - Yaoguang Zhang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, People's Republic of China.
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
- Beijing Hospital Continence Center, Beijing, People's Republic of China.
| |
Collapse
|
21
|
Wen T, Zhang Z. Cellular mechanisms of fibrin (ogen): insight from neurodegenerative diseases. Front Neurosci 2023; 17:1197094. [PMID: 37529232 PMCID: PMC10390316 DOI: 10.3389/fnins.2023.1197094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Neurodegenerative diseases are prevalent and currently incurable conditions that progressively impair cognitive, behavioral, and psychiatric functions of the central or peripheral nervous system. Fibrinogen, a macromolecular glycoprotein, plays a crucial role in the inflammatory response and tissue repair in the human body and interacts with various nervous system cells due to its unique molecular structure. Accumulating evidence suggests that fibrinogen deposits in the brains of patients with neurodegenerative diseases. By regulating pathophysiological mechanisms and signaling pathways, fibrinogen can exacerbate the neuro-pathological features of neurodegenerative diseases, while depletion of fibrinogen contributes to the amelioration of cognitive function impairment in patients. This review comprehensively summarizes the molecular mechanisms and biological functions of fibrinogen in central nervous system cells and neurodegenerative diseases, including Alzheimer's disease, Multiple Sclerosis, Parkinson's disease, Vascular dementia, Huntington's disease, and Amyotrophic Lateral Sclerosis. Additionally, we discuss the potential of fibrinogen-related treatments in the management of neurodegenerative disorders.
Collapse
|
22
|
Teller N, Chad JA, Wong A, Gunraj H, Ji X, Goubran M, Gilboa A, Roudaia E, Sekuler A, Churchill N, Schweizer T, Gao F, Masellis M, Lam B, Heyn C, Cheng I, Fowler R, Black SE, MacIntosh BJ, Graham SJ, Chen JJ. Feasibility of diffusion-tensor and correlated diffusion imaging for studying white-matter microstructural abnormalities: Application in COVID-19. Hum Brain Mapp 2023; 44:3998-4010. [PMID: 37162380 PMCID: PMC10258529 DOI: 10.1002/hbm.26322] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 05/11/2023] Open
Abstract
There has been growing attention on the effect of COVID-19 on white-matter microstructure, especially among those that self-isolated after being infected. There is also immense scientific interest and potential clinical utility to evaluate the sensitivity of single-shell diffusion magnetic resonance imaging (MRI) methods for detecting such effects. In this work, the performances of three single-shell-compatible diffusion MRI modeling methods are compared for detecting the effect of COVID-19, including diffusion-tensor imaging, diffusion-tensor decomposition of orthogonal moments and correlated diffusion imaging. Imaging was performed on self-isolated patients at the study initiation and 3-month follow-up, along with age- and sex-matched controls. We demonstrate through simulations and experimental data that correlated diffusion imaging is associated with far greater sensitivity, being the only one of the three single-shell methods to demonstrate COVID-19-related brain effects. Results suggest less restricted diffusion in the frontal lobe in COVID-19 patients, but also more restricted diffusion in the cerebellar white matter, in agreement with several existing studies highlighting the vulnerability of the cerebellum to COVID-19 infection. These results, taken together with the simulation results, suggest that a significant proportion of COVID-19 related white-matter microstructural pathology manifests as a change in tissue diffusivity. Interestingly, different b-values also confer different sensitivities to the effects. No significant difference was observed in patients at the 3-month follow-up, likely due to the limited size of the follow-up cohort. To summarize, correlated diffusion imaging is shown to be a viable single-shell diffusion analysis approach that allows us to uncover opposing patterns of diffusion changes in the frontal and cerebellar regions of COVID-19 patients, suggesting the two regions react differently to viral infection.
Collapse
Affiliation(s)
- Nick Teller
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
| | - Jordan A Chad
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Alexander Wong
- Department of System Design Engineering, University of Waterloo, Waterloo, Canada
| | - Hayden Gunraj
- Department of System Design Engineering, University of Waterloo, Waterloo, Canada
| | - Xiang Ji
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Maged Goubran
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Asaf Gilboa
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
- Department of Psychology, University of Toronto, Toronto, Canada
| | - Eugenie Roudaia
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
| | - Allison Sekuler
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
- Department of Psychology, University of Toronto, Toronto, Canada
| | - Nathan Churchill
- Neuroscience Research Program, St. Michael's Hospital, Toronto, Canada
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada
- Department of Physics, Toronto Metropolitan University, Toronto, Canada
| | - Tom Schweizer
- Neuroscience Research Program, St. Michael's Hospital, Toronto, Canada
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Canada
- Department of Neurosurgery, University of Toronto, Toronto, Canada
| | - Fuqiang Gao
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Mario Masellis
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Benjamin Lam
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Chris Heyn
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Ivy Cheng
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Robert Fowler
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Sandra E Black
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Bradley J MacIntosh
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - Simon J Graham
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, Canada
| | - J Jean Chen
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
23
|
Gao L, Pan X, Zhang JH, Xia Y. Glial cells: an important switch for the vascular function of the central nervous system. Front Cell Neurosci 2023; 17:1166770. [PMID: 37206667 PMCID: PMC10188976 DOI: 10.3389/fncel.2023.1166770] [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: 02/15/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
In this review, we first describe the current understanding of glial-mediated vascular function affecting the role of the blood-brain barrier (BBB) in central nervous system (CNS) disorders. BBB, mainly composed of glial and endothelial cells (ECs), is the protective structure that orchestrates the transport of substances, including ions, molecules, and cells from brain vessels into or out of the CNS. Then, we display the multiple communication between glial and vascular function based on angiogenesis, vascular wrapping, and blood perfusion in the brain. Glial can support microvascular ECs to form a blood network connecting to neurons. Astrocytes, microglia, and oligodendrocytes are the common types of glial surrounding the brain vessel. Glial-vessel interaction is required for the permeability and integrity of BBB. Glial cells surrounding the cerebral blood vessels can transmit communication signals to ECs and regulate the activity of vascular endothelial growth factor (VEGF) or Wnt-dependent endothelial angiogenesis mechanism. In addition, these glial cells monitor the blood flow in the brain via Ca2+/K+-dependent pathways. Finally, we provide a potential research direction for the glial-vessel axis in CNS disorders. Microglial activation can trigger astrocyte activation, which suggests that microglia-astrocyte interaction may play a key role in monitoring cerebral blood flow. Thus, microglia-astrocyte interaction can be the key point of follow-up studies focusing on the microglia-blood mechanism. More investigations focus on the mechanism of how oligodendrocyte progenitor cells communicate and interact with ECs. The direct role of oligodendrocytes in modulating vascular function needs to be explored in the future.
Collapse
Affiliation(s)
- Ling Gao
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Xuezhen Pan
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
| | - John H. Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
| |
Collapse
|
24
|
McLarnon JG. Glial-derived Neuroinflammation induced with Amyloid-beta-peptide Plus Fibrinogen Injection in Rat Hippocampus. Curr Alzheimer Res 2023; 20:515-522. [PMID: 37702232 DOI: 10.2174/1567205020666230912113501] [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: 05/23/2023] [Revised: 06/27/2023] [Accepted: 08/08/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION The present study has examined microglial and astrocyte activation in association with neuronal degeneration in an animal model using an injection of amyloid-beta peptide Aβ1-42 (Aβ42) plus fibrinogen into rat hippocampus. METHODS The combination of stimuli is suggested as a novel and potent perturbation to induce gliosis and the production of glial-derived neurotoxic factors in an animal model exhibiting a leaky BBB (blood-brain barrier). Specifically, Aβ42 + fibrinogen stimulation elevated levels of COX-2 (cyclooxygenase-2) and iNOS (inducible nitric oxide synthase) with a considerable extent of neuronal loss associated with microglia and astrocyte activation. RESULTS Treatment of injected rats with the broad spectrum anti-inflammatory agent, minocycline or the iNOS inhibitor, 1400 W inhibited gliosis, reduced levels of COX-2 and iNOS, and demonstrated efficacy for neuroprotection. CONCLUSION The findings suggest the utility of combining amyloid beta peptide plus fibrinogen as a potent and understudied neuroinflammatory stimulus for the induction of glial-derived neurotoxic factors in BBB-compromised AD brain.
Collapse
Affiliation(s)
- James G McLarnon
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, The University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
| |
Collapse
|
25
|
Bian Z, Hu X, Liu X, Yu H, Bian Y, Sun H, Fukui Y, Morihara R, Ishiura H, Yamashita T. Protective Effects of Rivaroxaban on White Matter Integrity and Remyelination in a Mouse Model of Alzheimer's Disease Combined with Cerebral Hypoperfusion. J Alzheimers Dis 2023; 96:609-622. [PMID: 37840489 DOI: 10.3233/jad-230413] [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] [Indexed: 10/17/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cognitive dysfunction and memory loss that is accompanied by pathological changes to white matter. Some clinical and animal research revealed that AD combined with chronic cerebral hypoperfusion (CCH) exacerbates AD progression by inducing blood-brain barrier dysfunction and fibrinogen deposition. Rivaroxaban, an anticoagulant, has been shown to reduce the rates of dementia in atrial fibrillation patients, but its effects on white matter and the underlying mechanisms are unclear. OBJECTIVE The main purpose of this study was to explore the therapeutic effect of rivaroxaban on the white matter of AD+CCH mice. METHODS In this study, the therapeutic effects of rivaroxaban on white matter in a mouse AD+CCH model were investigated to explore the potential mechanisms involving fibrinogen deposition, inflammation, and oxidative stress on remyelination in white matter. RESULTS The results indicate that rivaroxaban significantly attenuated fibrinogen deposition, fibrinogen-related microglia activation, oxidative stress, and enhanced demyelination in AD+CCH mice, leading to improved white matter integrity, reduced axonal damage, and restored myelin loss. CONCLUSIONS These findings suggest that long-term administration of rivaroxaban might reduce the risk of dementia.
Collapse
Affiliation(s)
- Zhihong Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xia Liu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Yu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yuting Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hongming Sun
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hiroyuki Ishiura
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| |
Collapse
|
26
|
Duan K, Ma Y, Tan J, Miao Y, Zhang Q. Identification of genetic molecular markers and immune infiltration characteristics of Alzheimer's disease through weighted gene co-expression network analysis. Front Neurol 2022; 13:947781. [PMID: 36071897 PMCID: PMC9441600 DOI: 10.3389/fneur.2022.947781] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Background Alzheimer's disease (AD) is a progressive neurodegenerative disease that leads to cognitive impairment and memory loss. Currently, the pathogenesis and underlying causative genes of AD remain unclear, and there exists no effective treatment for this disease. This study explored AD-related diagnostic and therapeutic biomarkers from the perspective of immune infiltration by analyzing public data from the NCBI Gene Expression Omnibus database. Method In this study, weighted gene co-expression network analysis (WGCNA) was conducted to identify modules and hub genes contributing to AD development. A protein–protein interaction network was constructed when the genes in the modules were enriched and examined by Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Furthermore, a gene network was established using topological WGCNA, from which five hub genes were selected. Logistic regression analysis and receiver operating characteristic curve analysis were performed to explore the clinical value of genes in AD diagnosis. The genes in the core module intersected with the hub genes, and four intersection genes (ATP2A2, ATP6V1D, CAP2, and SYNJ1) were selected. These four genes were enriched by gene set enrichment analysis (GSEA). Finally, an immune infiltration analysis was performed. Results The GO/KEGG analysis suggested that genes in the core module played a role in the differentiation and growth of neural cells and in the transmission of neurotransmitters. The GSEA of core genes showed that these four genes were mainly enriched in immune/infection pathways (e.g., cholera infection and Helicobacter pylori infection pathways) and other metabolic pathways. An investigation of immune infiltration characteristics revealed that activated mast cells, regulatory T cells, plasma cells, neutrophils, T follicular helper cells, CD8 T cells, resting memory CD4 T cells, and M1 macrophages were the core immune cells contributing to AD progression. qRT-PCR analysis showed that the ATP6V1D is upregulated in AD. Conclusion The results of enrichment and immuno-osmotic analyses indicated that immune pathways and immune cells played an important role in the occurrence and development of AD. The selected key genes were used as biomarkers related to the pathogenesis of AD to further explore the pathways and cells, which provided new perspectives on therapeutic targets in AD.
Collapse
Affiliation(s)
- KeFei Duan
- Department of Geriatrics, Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuan Ma
- Department of Geriatrics, Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuyang Miao
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Qiang Zhang
- Department of Geriatrics, Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Qiang Zhang
| |
Collapse
|
27
|
Hao Y, Li J, Yue S, Wang S, Hu S, Li B. Neuroprotective Effect and Possible Mechanisms of Berberine in Diabetes-Related Cognitive Impairment: A Systematic Review and Meta-Analysis of Animal Studies. Front Pharmacol 2022; 13:917375. [PMID: 35734409 PMCID: PMC9208278 DOI: 10.3389/fphar.2022.917375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/02/2022] [Indexed: 12/09/2022] Open
Abstract
Berberine, the main bioactive component of Coptis chinensis Franch., is widely used in the treatment of diabetes. Previous studies have reported that berberine supplementation may play a multitarget therapeutic role in diabetes-related cognitive impairment (DCI). This systematic review and meta-analysis evaluated the effect and possible mechanisms of berberine in animal models of DCI. Relevant studies were searched through PubMed, Web of Science, Embase, and three Chinese databases (CNKI, Wanfang, and VIP) until March 2022. Twenty studies involving 442 animals were included, and SYRCLE’s risk of bias tool was used to assess methodological quality. The statistical analysis was performed using STATA 15.0 to calculate the weighted standard mean difference (SMD) with a 95% confidence interval (CI). The fasting blood glucose (FBG) and Morris water maze test (MWM) were the main outcomes to be analyzed. The overall results showed that berberine could significantly improve FBG, escape latency, the times of crossing the platform, the time spent in the target quadrant, serum insulin, 2hBG of oral glucose tolerance test (OGTT), amyloid β (Aβ), acetylcholinesterase (AChE), oxidative stress, and inflammation levels. The present meta-analysis demonstrated that berberine could not only lower blood glucose levels but also improve learning and memory in DCI animal models, which might involve regulating glucose and lipid metabolism, improving insulin resistance, anti-oxidation, anti-neuroinflammation, inhibiting endoplasmic reticulum (ER) stress; and improving the cholinergic system. However, additional attention should be paid to these outcomes due to the significant heterogeneity.
Collapse
Affiliation(s)
- Yanwei Hao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiaxin Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shengnan Yue
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaofeng Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuangyuan Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Bin Li,
| |
Collapse
|
28
|
Yoshikawa M, Aizawa S, Oppenheim RW, Milligan C. Neurovascular unit pathology is observed very early in disease progression in the mutant SOD1G93A mouse model of amyotrophic lateral sclerosis. Exp Neurol 2022; 353:114084. [DOI: 10.1016/j.expneurol.2022.114084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 11/26/2022]
|