1
|
Shi Z, Kuai M, Li B, Akowuah CF, Wang Z, Pan Y, Tang M, Yang X, Lü P. The role of VEGF in Cancer angiogenesis and tumorigenesis: Insights for anti-VEGF therapy. Cytokine 2025; 189:156908. [PMID: 40049050 DOI: 10.1016/j.cyto.2025.156908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/16/2025] [Accepted: 02/28/2025] [Indexed: 03/18/2025]
Abstract
Vascular endothelial growth factor (VEGF) is a critical regulator of angiogenesis, playing a pivotal role in both physiological and pathological processes. It promotes the formation of new blood vessels and activates downstream signaling pathways that regulate endothelial cell function. This review highlights recent advancements in the understanding of VEGF's molecular structure and its isoforms, as well as their implications in disease progression. It also explores the mechanisms of VEGF inhibitors. While VEGF inhibitors show promise in the treatment of cancer and other diseases, their clinical use faces significant challenges, including drug resistance, side effects, and complex interactions with other signaling pathways. To address these challenges, future research should focus on: (i) enhancing the understanding of VEGF subtypes and their distinct roles in various diseases, supporting the development of personalized treatment strategies; (ii) developing combination therapies that integrate VEGF inhibitors with other targeted treatments to overcome resistance and improve efficacy; (iii) optimizing drug delivery systems to reduce off-target effects and enhance therapeutic outcomes. These approaches aim to improve the effectiveness and safety of VEGF-targeted therapies, offering new possibilities for the treatment of VEGF-related diseases.
Collapse
Affiliation(s)
- Zijun Shi
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Mengmeng Kuai
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Baohua Li
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | | | - Zhenyu Wang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Ye Pan
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoyue Yang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China.
| | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China; Affiliated Hospital of Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
2
|
Ghanbarian E, Khorsand B, Petersen KK, Nallapu BT, Sajjadi SA, Lipton RB, Ezzati A. Cerebrospinal fluid inflammatory cytokines as prognostic indicators for cognitive decline across Alzheimer's disease spectrum. J Alzheimers Dis 2025:13872877251335915. [PMID: 40262111 DOI: 10.1177/13872877251335915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BackgroundNeuroinflammation actively contributes to the pathophysiology of Alzheimer's disease (AD); however, the value of neuroinflammatory biomarkers for disease-staging or predicting disease progression remains unclear.ObjectiveTo investigate diagnostic and prognostic utility of inflammatory biomarkers in combination with conventional AD biomarkers.MethodsData from 258 participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI) with cerebrospinal fluid (CSF) biomarkers of amyloid-β (Aβ), tau, and inflammation were analyzed. Clinically meaningful cognitive decline (CMCD) was defined as a ≥ 4-point increase on the Alzheimer's Disease Assessment Scale Cognitive Subscore 11. Predictor variables included demographics (D: age, sex, education), APOE4 status (A), inflammatory biomarkers (I), and classic AD biomarkers of Aβ and p-tau181 (C). Models incorporating inflammatory biomarkers assessed their contribution to improving baseline diagnostic classification and 1-year CMCD prediction.ResultsAt 1-year follow-up, 27.1% of participants experienced CMCD. Adding inflammatory biomarkers to models with D and A variables (DA model) improved classification of cognitively normal (CN) versus mild cognitive impairment (MCI) and CN versus Dementia (p < 0.001). Similarly, inflammatory markers enhanced classification in models including C (DAC model), for CN versus MCI (p < 0.01) and CN versus Dementia (p < 0.001). Predictive performance for CMCD was improved in individuals with MCI and dementia in both models (all p < 0.05). In addition, the DAI model outperformed the DAC model in predicting CMCD for MCI and Dementia groups (both p < 0.05).ConclusionsAddition of CSF inflammatory biomarkers to biomarkers of AD improves diagnostic accuracy of clinical disease stage at baseline and add incremental value to AD biomarkers for prediction of cognitive decline.
Collapse
Affiliation(s)
- Elham Ghanbarian
- Department of Neurology, University of California, Irvine, CA, USA
| | - Babak Khorsand
- Department of Neurology, University of California, Irvine, CA, USA
| | - Kellen K Petersen
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Bhargav T Nallapu
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - S Ahmad Sajjadi
- Department of Neurology, University of California, Irvine, CA, USA
| | - Richard B Lipton
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Ali Ezzati
- Department of Neurology, University of California, Irvine, CA, USA
| |
Collapse
|
3
|
Jaime Garcia D, Clancy U, Arteaga C, Valdés-Hernandez MC, Chappell FM, Jochems ACC, Cheng Y, Zhang J, Thrippleton MJ, Stringer MS, Sleight E, Backhouse EV, Wiseman S, Brown R, Doubal FN, Montagne A, Wardlaw JM. Blood biomarkers of vascular dysfunction in small vessel disease progression: Insights from a longitudinal neuroimaging study. Alzheimers Dement 2025; 21:e70152. [PMID: 40275856 DOI: 10.1002/alz.70152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 04/26/2025]
Abstract
INTRODUCTION This study explored the relationship between blood biomarkers of cerebrovascular function and small vessel disease (SVD) neuroimaging markers and cognitive outcomes in highly-phenotyped participants. METHODS We conducted cross-sectional and 1-year longitudinal analyses on 181 patients with mild ischemic stroke, enriched for SVD features. We examined relationships between a panel of 13 blood biomarkers and magnetic resonance imaging (MRI) markers of SVD (structural lesions, diffusion-weighted imaging [DWI]-positive lesions, blood-brain barrier (BBB) permeability, and cerebrovascular reactivity (CVR), and cognition. RESULTS In linear mixed models, vascular endothelial growth factor was significantly associated with incident DWI-positive lesions over 1 year. Intercellular adhesion molecule-1 was linked with lower CVR while platelet-derived growth factor-subunit B and Endothelin-1 were associated with higher CVR. Platelet-Selectin levels were associated with mild cognitive impairment at 1 year. DISCUSSION Our results support the role of endothelial and pericyte dysfunction in SVD burden and progression and suggest that specific biomarkers relate to distinct SVD manifestations. HIGHLIGHTS Small vessel disease (SVD) lacks specific or predictive biomarker signatures. Vascular endothelial growth factor levels were linked to incident lesions detected over 1 year. Circulating intercellular adhesion molecule-1 related to lower cerebrovascular reactivity. Platelet-selectin levels were associated with mild cognitive impairment longitudinally. These findings could help stratify patients at high-risk of rapid-progression SVD.
Collapse
Affiliation(s)
- Daniela Jaime Garcia
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - Una Clancy
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Carmen Arteaga
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Maria C Valdés-Hernandez
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - Francesca M Chappell
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Angela C C Jochems
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - Yajun Cheng
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Junfang Zhang
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Michael J Thrippleton
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility (Royal Infirmary of Edinburgh), University of Edinburgh, Edinburgh, UK
| | - Michael S Stringer
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility (Royal Infirmary of Edinburgh), University of Edinburgh, Edinburgh, UK
| | - Emilie Sleight
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - Ellen V Backhouse
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - Stewart Wiseman
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - Rosalind Brown
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - Fergus N Doubal
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Axel Montagne
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, School of Clinical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Peng X, Mei Z, Luo Z, Ge J. Stroke with White Matter Lesions: Potential Pathophysiology and Therapeutic Targets. Br J Hosp Med (Lond) 2025; 86:1-21. [PMID: 40135304 DOI: 10.12968/hmed.2024.0771] [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: 03/27/2025]
Abstract
Stroke is one of the most common causes of morbidity and mortality among adults globally. Significant advancements have been made in elucidating its pathophysiology, with stroke categorized into pathological subtypes, such as ischemic stroke (IS) and hemorrhagic stroke. White matter lesions (WMLs) identified on magnetic resonance imaging rank as a hallmark of cerebral small vessel disease and are associated with vascular risk factors. They are linked to adverse outcomes like dementia, depression, and an increased risk of both first-ever and recurrent strokes, independent of other risk factors. Despite the evidence indicating the close link between WMLs and stroke, their underlying pathophysiological relationship remains unclear. This study aims to provide an overview of the current knowledge and recent advances in epidemiology, risk factors, and pathophysiological mechanisms of WMLs and stroke, focusing on their interconnection and emerging therapeutic targets.
Collapse
Affiliation(s)
- Xiwen Peng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhenghua Luo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Spinal Department, The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
5
|
Zoccali C, Mallamaci F, Wagner CA, Unwin R, Nedergaard M, Hafez G, Malyszko J, Pepin M, Massy Z, Paolisso G, Remuzzi G, Capasso GB. Genetic and circulating biomarkers of cognitive dysfunction and dementia in CKD. Nephrol Dial Transplant 2025; 40:ii64-ii75. [PMID: 40080085 PMCID: PMC11905751 DOI: 10.1093/ndt/gfae259] [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: 07/15/2024] [Indexed: 03/15/2025] Open
Abstract
Chronic kidney disease (CKD) is commonly accompanied by cognitive dysfunction and dementia, which, in turn, increase the risk of hospitalization, cardiovascular events and death. Over the last 30 years, only four studies focused on genetic markers of cognitive impairment in CKD and kidney failure (KF), indicating a significant gap in research. These studies suggest potential genetic predispositions to cognitive decline in CKD patients but also underscore the necessity for more comprehensive studies. Seventeen reports have established connections between cognitive function and kidney disease markers such as estimated glomerular filtration rate (eGFR), Cystatin C and albuminuria. A rapid eGFR decline has been associated with cognitive deterioration and vascular dementia, and mild to moderate eGFR reductions with diminished executive function in elderly men. Various biomarkers have been associated to Alzheimer's disease or dementia in CKD and KF. These include amyloid beta and phosphorylated tau proteins, uremic toxins, gut microbiota, metabolic indicators, hypertension, endothelial dysfunction, vitamins and inflammation. However, the causal relevance of these associations remains unclear. Overall, the available evidence points to a complex interplay between the different biomarkers and cognitive health in CKD patients, underscoring the need for more research to elucidate these relationships.
Collapse
Affiliation(s)
- Carmine Zoccali
- Renal Research Institute, NY, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Francesca Mallamaci
- Nephrology, Dialysis and Transplantation Unit, Grande Ospedale Metropolitano
- CNR-IFC, Institute of Clinical Physiology, Research Unit of Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension of Reggio Calabria, Italy
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Robert Unwin
- UCL Department of Renal Medicine, Royal Free Hospital, London, UK
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gaye Hafez
- Department of Pharmacology, Faculty of Pharmacy, Altinbas University, Istanbul, Turkey
| | - Jolanta Malyszko
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Marion Pepin
- Department of Nephrology, Ambroise Paré University Medical Center, APHP, Paris, France
- Department of Geriatrics, Ambroise Paré University Medical Center, APHP, Boulogne-Billancourt, France
| | - Ziad Massy
- Paris-Saclay University, UVSQ, Inserm, Clinical Epidemiology Team, Centre de Recherche en Epidémiologie et Santé des Populations (CESP), Villejuif, France
- Association pour l'Utilisation du Rein Artificiel (AURA), Paris and Department of Nephrology, Ambroise Paré University Medical Center, APHP, Paris, France
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- UniCamillus, International Medical University, Rome, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Giovambattista B Capasso
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
6
|
Jiang YQ, Chen QZ, Yang Y, Zang CX, Ma JW, Wang JR, Dong YR, Zhou N, Yang X, Li FF, Bao XQ, Zhang D. White matter lesions contribute to motor and non-motor disorders in Parkinson's disease: a critical review. GeroScience 2025; 47:591-609. [PMID: 39576561 PMCID: PMC11872850 DOI: 10.1007/s11357-024-01428-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/08/2024] [Indexed: 03/04/2025] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease, characterized by movement disorders and non-motor symptoms like cognitive impairment and depression. Degeneration of dopaminergic neurons in the substantia nigra and Lewy bodies have long been considered as main neuropathological changes. However, recent magnetic resonance imaging (MRI) studies have shown that white matter lesions (WMLs) were present in PD patients. WMLs are characterized by loss or impairment of myelin sheath in central nerve fibers, which are closely correlated with motor and cognitive dysfunction in PD. WMLs alterations precede nigrostriatal neuronal losses and can independently affect the clinical severity or characteristics of motor coordination in PD patients. Currently, the exact mechanism of WMLs involvement in the occurrence and development of PD remains unclear. It is speculated that WMLs may participate in the pathogenesis of PD by disrupting important connections in brain or promoting axonal degeneration. In this review, we will discuss the pathological changes and mechanisms of WMLs, elaborate the impact of WMLs on the progression of PD, clarify the importance of WMLs in PD pathogenesis, and thus provide novel targets for PD treatments.
Collapse
Affiliation(s)
- Yue-Qi Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Qiu-Zhu Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Yang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Cai-Xia Zang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Jing-Wei Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Jin-Rong Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Yi-Rong Dong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Ning Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Xing Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Fang-Fang Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Xiu-Qi Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China.
| | - Dan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China.
| |
Collapse
|
7
|
Gao F, Zhan Y, Wang Q, Zhang M, Dai L, Shen Y. Pathological angiogenesis was associated with cerebrovascular lesion and neurodegeneration in Alzheimer's disease. Alzheimers Dement 2025; 21:e14521. [PMID: 39777972 PMCID: PMC11848169 DOI: 10.1002/alz.14521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/24/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION We investigated the specific factors driving abnormal angiogenesis in Alzheimer's disease (AD) and its role in cerebrovascular lesions and neurodegeneration. METHODS We assessed cerebrovascular pathologies, amyloid-beta (Aβ), and tau pathologies in post mortem human brains and detected 12 angiogenic factors in cerebrospinal fluid (CSF) from the China Aging and Neurodegenerative Disease Initiative (CANDI) cohort. RESULTS We observed severe blood-brain barrier damage and elevated levels of the vascular marker CD31 in human AD brains, which had a stronger correlation with tau pathology than Aβ pathology. Consistently, only CSF pTau181 showed positive associations with CSF angiogenesis factors (soluble vascular endothelial growth factor receptor 2 [sVEGFR2], VEGF-C, VEGF-D, placental growth factor [PLGF], Angiopoietin2, and Serpin E1), but not CSF Aβ42/40. Additionally, higher levels of CSF sVEGFR1, soluble Tyrosine-protein kinase receptor 2 [sTIE2], PLGF, and interleukin 8 [IL8], as well as lower levels of CSF urokinase-type plasminogen activator [uPA], were associated with worsen cerebrovascular pathologies and neurodegeneration. DISCUSSION Our findings indicate that tau pathology may play a critical role in pathological angiogenesis, contributing to cerebrovascular lesions and neurodegeneration in AD. HIGHLIGHTS BBB damage and elevated vascular marker CD31 in human AD brains had a stronger correlation with tau pathology than Aβ pathology. CSF pTau181 mediated the effect of CSF Aβ42/40 on CSF sVEGFR1 and sTIE2. Only CSF pTau181 showed positive associations with sVEGFR2, VEGF-C, VEGF-D, PLGF, Angiopoietin2, and Serpin E1. Higher sVEGFR1, sTIE2, PLGF, and IL8, and lower uPA in CSF, were associated with cerebrovascular pathologies and neurodegeneration.
Collapse
Affiliation(s)
- Feng Gao
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Yaxi Zhan
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Qiong Wang
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Mengguo Zhang
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Linbin Dai
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Yong Shen
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
8
|
Iadecola C, Anrather J. The immunology of stroke and dementia. Immunity 2025; 58:18-39. [PMID: 39813992 PMCID: PMC11736048 DOI: 10.1016/j.immuni.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Ischemic stroke and vascular cognitive impairment, caused by a sudden arterial occlusion or more subtle but protracted vascular insufficiency, respectively, are leading causes of morbidity and mortality worldwide with limited therapeutic options. Innate and adaptive immunity have long been implicated in neurovascular injury, but recent advances in methodology and new experimental approaches have shed new light on their contributions. A previously unappreciated dynamic interplay of brain-resident, meningeal, and systemic immune cells with the ischemic brain and its vasculature has emerged, and new insights into the frequent overlap between vascular and Alzheimer pathology have been provided. Here, we critically review these recent findings, place them in the context of current concepts on neurovascular pathologies and Alzheimer's disease, and highlight their impact on recent stroke and Alzheimer therapies.
Collapse
Affiliation(s)
- Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
9
|
Qiao Y, Zhao L, Cong C, Li Y, Tian S, Zhu X, Yang J, Cao S, Li P, Su J. Association of systemic inflammatory markers with white matter hyperintensities and microstructural injury: an analysis of UK Biobank data. J Psychiatry Neurosci 2025; 50:E45-E56. [PMID: 39848683 PMCID: PMC11771994 DOI: 10.1503/jpn.240039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/30/2024] [Accepted: 12/07/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND White matter damage is closely associated with cognitive and psychiatric symptoms and is prevalent in cerebral small vessel disease (CSVD); although the pathophysiological mechanisms involved in CSVD remain elusive, inflammation plays a crucial role. We sought to investigate the relationship between systemic inflammation markers and imaging markers of CVSD, namely white matter hyperintensity (WMH) and microstructural injury. METHODS We conducted a study involving both cross-sectional and longitudinal data from the UK Biobank Cohort. We performed multiple linear regression analyses, adjusted for potential confounders, to explore the associations between systemic inflammation markers (e.g., systemic immune-inflammation index [SII], neutrophil-to-lymphocyte ratio [NLR], C-reactive protein [CRP] levels, monocyte count, neutrophil count) and macro- and microstructural white matter injury, as markers of CSVD. We performed Mendelian randomization analysis to investigate the genetically predictive effect of monocytes on WMH, as well as mediation analysis to clarify whether inflammatory markers affected cognitive function via white matter injury. RESULTS We included 36 411 participants (mean age 54.8 ± 7.5 yr, 51.9% female) from the UK Biobank Cohort. We found that SII was significantly associated with both WMH and microstructural injury markers (fractional anisotropy, mean diffusivity, intracellular volume fraction, and isotropic compartment volume fraction [ISOVF]), and the neutrophil-to-lymphocyte ratio was significantly associated with WMH and some markers of microstructural injury (mean diffusivity and ISOVF). Our analysis revealed that the CRP level was significantly associated with WMH and WMH progression but not with microstructural injury. We also demonstrated that monocyte count was significantly associated with WMH and ISOVF, and that neutrophil count was significantly associated with WMH, mean diffusivity, and ISOVF. In 2-sample Mendelian randomization analyses, we found positive associations between genetic determinants of monocytes and WMH. The mediating role of WMH suggested that a higher SII value and monocyte count could contribute to cognitive impairment through white matter injury. LIMITATIONS Although the study includes both cross-sectional and longitudinal components, the sample size for the longitudinal aspect is limited, and the use of blood biomarkers from a single timepoint is also a limitation of this research. CONCLUSION The SII and neutrophil-to-lymphocyte ratio may be early detection markers for white matter damage in patients with CSVD, whereas the CRP level is more closely associated with disease severity and progression. Our findings highlight the clinical relevance of systemic inflammation markers with white matter macro- and microstructural injuries, revealing that systemic inflammation is likely involved in the mechanism of early white matter injury among patients with CSVD.
Collapse
Affiliation(s)
- Yuan Qiao
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| | - Lei Zhao
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| | - Chaohua Cong
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| | - Yuna Li
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| | - Shan Tian
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| | - Xirui Zhu
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| | - Junting Yang
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| | - Shanshan Cao
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| | - Panlong Li
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| | - Jingjing Su
- From the Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Qiao, Zhao, Cong, Y. Li, Tian, Yang, Cao, Su); the School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China (Zhu); the Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China (P. Li)
| |
Collapse
|
10
|
Righi D, Manco C, Pardini M, Stufano A, Schino V, Pelagotti V, Massa F, Stefano ND, Plantone D. Investigating interleukin-8 in Alzheimer's disease: A comprehensive review. J Alzheimers Dis 2025; 103:38-55. [PMID: 39558604 DOI: 10.1177/13872877241298973] [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: 11/20/2024]
Abstract
Several studies indicate that the development of Alzheimer's disease (AD) has strong interactions with immune mechanisms within the brain, indicating a close association between inflammation in the central nervous system and the progression of neurodegeneration. Despite considerable progress in understanding the inflammatory aspects of AD, several of them remain unresolved. Pro-inflammatory cytokines and microglia are pivotal components in the inflammatory cascade. Among these, the role of interleukin-8 (IL-8) in neurodegeneration seems complex and multifaceted, involving inflammation, neurotoxicity, blood-brain barrier disruption, and oxidative stress, and is still poorly characterized. We conducted a review to describe the evidence of IL-8 involvement in AD. IL-8 is a cytokine known for its proinflammatory properties and typically produced by macrophages, predominantly functions as a chemotactic signal for attracting neutrophils to inflamed sites in the bloodstream. Interestingly, IL-8 is also present in the brain, where it is primarily released by microglia in response to inflammatory signals. This review aims to provide a comprehensive overview of the structure, function, and regulatory mechanisms of IL-8 relevant to AD pathology.
Collapse
Affiliation(s)
- Delia Righi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Carlo Manco
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Angela Stufano
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Valentina Schino
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Virginia Pelagotti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Domenico Plantone
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
11
|
Cox JG, Cole JH, Kempton MJ, Williams SCR, de Groot M. Volume and distribution of white matter hyperintensities in rheumatoid arthritis and ulcerative colitis patients. Sci Rep 2024; 14:32010. [PMID: 39738366 PMCID: PMC11685908 DOI: 10.1038/s41598-024-83559-1] [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: 05/30/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
Brain white matter disruptions have been implicated in contributing to fatigue, brain fog and other central symptoms commonly reported in inflammatory diseases. In this study, we included 252 RA patients with 756 age and sex matched controls and 240 UC patients with 720 age and sex matched controls using the UK Biobank imaging dataset. We looked for differences in total volume of white matter hyperintensities (WMH) between patients compared to controls. Then, using voxelwise analysis, we explored the spatial distribution of these white matter hyperintensities and differences in these between patients and controls and between disease groups. A significantly higher volume of WMH was observed in both the RA (p = 1.9 × 10-8, β = - 0.36, 95% CI = - 0.48, - 0.23) and UC (p = 0.003, β = - 0.18 95% CI = - 0.31, - 0.06) patients compared to their respective control groups. Voxelwise analysis revealed only a small cluster of RA associated WMH compared to controls. These results indicate an increased risk of white matter hyperintensities in patients with RA and UC. These findings help quantify the effect of inflammation from autoimmune diseases on cerebrovascular health and white matter integrity.
Collapse
Affiliation(s)
- Jennifer G Cox
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - James H Cole
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - Matthew J Kempton
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Steven C R Williams
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marius de Groot
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Groover Consulting, Rotterdam, The Netherlands
| |
Collapse
|
12
|
Wang Q, Yuan S, Wang C, Huang D, Zhang M, Zhan Y, Gao F, Shi J, Levey AI, Shen Y. Brain derived β-interferon is a potential player in Alzheimer's disease pathogenesis and cognitive impairment. Alzheimers Res Ther 2024; 16:271. [PMID: 39709485 DOI: 10.1186/s13195-024-01644-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Recent research has postulated that the activation of cGAS-STING-interferon signalling pathways could be implicated in the pathogenesis of Alzheimer's disease (AD). However, the precise types of interferons and related cytokines, both from the brain and periphery, responsible for cognitive impairment in patients with AD remain unclear. METHODS A total of 131 participants (78 [59.5%] female and 53 [40.5%] male; mean [SD] age, 61.5 [7.6] years) with normal cognition and cognitive impairment from the China Aging and Neurodegenerative Initiative cohort were included. CSF and serum IFNα-2a, IFN-β, IFN-γ, TNF-α, IL-6, IL-10, MCP-1and CXCL-10 were tested. The correlation between these interferons and related cytokines with AD core biomarkers in the CSF and plasma, cognition performance, and brain MRI measures were analysed. RESULTS We found that only CSF IFN-β levels were significantly elevated in Alzheimer's disease compared to normal cognition. Furthermore, CSF IFN-β levels were significantly associated with AD core biomarkers (CSF P-tau and Aβ42/Aβ40 ratio) and cognitive performance (MMSE and CDR score). Additionally, the CSF IFN-β levels were significantly correlated with the typical pattern of brain atrophy in AD (such as hippocampus, amygdala, and precuneus). In contrast, CSF IL-6 levels were significantly elevated in non-AD cognitively impaired patients compared to other groups. Moreover, CSF IL-6 levels were significantly associated with cognitive performance in non-AD individuals and correlated with the vascular cognitive impairment-related MRI markers (such as white matter hyperintensity). CONCLUSION Our findings demonstrate that distinct inflammatory molecules are associated with different cognitive disorders. Notably, CSF IFN-β levels are significantly linked to the pathology and cognitive performance of AD, identifying this interferon as a potential target for AD therapy.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Neurology and Institute on Aging and Brain Disorders, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Lujiang Road 17, Hefei, 230001, China.
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China.
| | - Shufen Yuan
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Chenxi Wang
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Duntao Huang
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Mengguo Zhang
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Yaxi Zhan
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Feng Gao
- Department of Neurology and Institute on Aging and Brain Disorders, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Lujiang Road 17, Hefei, 230001, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Jiong Shi
- Department of Neurology and Institute on Aging and Brain Disorders, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Lujiang Road 17, Hefei, 230001, China
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA
| | - Yong Shen
- Department of Neurology and Institute on Aging and Brain Disorders, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Lujiang Road 17, Hefei, 230001, China.
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China.
| |
Collapse
|
13
|
You T, Wang Y, Chen S, Dong Q, Yu J, Cui M. Vascular cognitive impairment: Advances in clinical research and management. Chin Med J (Engl) 2024; 137:2793-2807. [PMID: 39048312 PMCID: PMC11649275 DOI: 10.1097/cm9.0000000000003220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Indexed: 07/27/2024] Open
Abstract
ABSTRACT Vascular cognitive impairment (VCI) encompasses a wide spectrum of cognitive disorders, ranging from mild cognitive impairment to vascular dementia. Its diagnosis relies on thorough clinical evaluations and neuroimaging. VCI predominately arises from vascular risk factors (VRFs) and cerebrovascular disease, either independently or in conjunction with neurodegeneration. Growing evidence underscores the prevalence of VRFs, highlighting their potential for early prediction of cognitive impairment and dementia in later life. The precise mechanisms linking vascular pathologies to cognitive deficits remain elusive. Chronic cerebrovascular pathology is the most common neuropathological feature of VCI, often interacting synergistically with neurodegenerative processes. Current research efforts are focused on developing and validating reliable biomarkers to unravel the etiology of vascular brain changes in VCI. The collaborative integration of these biomarkers into clinical practice, alongside routine incorporation into neuropathological assessments, presents a promising strategy for predicting and stratifying VCI. The cornerstone of VCI prevention remains the control of VRFs, which includes multi-domain lifestyle modifications. Identifying appropriate pharmacological approaches is also of paramount importance. In this review, we synthesize recent advancements in the field of VCI, including its definition, determinants of vascular risk, pathophysiology, neuroimaging and fluid-correlated biomarkers, predictive methodologies, and current intervention strategies. Increasingly evident is the notion that more rigorous research for VCI, which arises from a complex interplay of physiological events, is still needed to pave the way for better clinical outcomes and enhanced quality of life for affected individuals.
Collapse
Affiliation(s)
- Tongyao You
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yingzhe Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shufen Chen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jintai Yu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200040, China
| |
Collapse
|
14
|
Hu M, Bai C, Zhao H, Wu J, Luan X. Research Progress on the Role of the Interleukin Family in the Pathogenesis of Cerebral Palsy in Children. J Integr Neurosci 2024; 23:213. [PMID: 39735959 DOI: 10.31083/j.jin2312213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 12/31/2024] Open
Abstract
Cerebral palsy (CP), a common neurological disorder in children, remains a significant research focus. The interleukin (IL) family, pivotal mediators in inflammatory responses, shows increased expression in various neuroinflammatory diseases, markedly influencing their onset and progression. Elevated IL levels in the brains of children with CP, in contrast to healthy peers, reflect similar elevations in neurological conditions linked to CP, indicating a strong association between CP and the IL family. Anti-inflammatory therapies, particularly those targeting ILs, have shown effectiveness in animal models, diverging from traditional CP management methods. This shift suggests IL modulation as a promising therapeutic strategy in pediatric CP. This review consolidates recent findings on the IL family's role in CP, illuminating their evolving relationship.
Collapse
Affiliation(s)
- Mingbo Hu
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| | - Chao Bai
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| | - Hong Zhao
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| | - Junjie Wu
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| | - Xinping Luan
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| |
Collapse
|
15
|
Svenningsson AL, Bocancea DI, Stomrud E, van Loenhoud A, Barkhof F, Mattsson-Carlgren N, Palmqvist S, Hansson O, Ossenkoppele R. Biological mechanisms of resilience to tau pathology in Alzheimer's disease. Alzheimers Res Ther 2024; 16:221. [PMID: 39396028 PMCID: PMC11470552 DOI: 10.1186/s13195-024-01591-9] [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: 02/14/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND In Alzheimer's disease (AD), the associations between tau pathology and brain atrophy and cognitive decline are well established, but imperfect. We investigate whether cerebrospinal fluid (CSF) biomarkers of biological processes (vascular, synaptic, and axonal integrity, neuroinflammation, neurotrophic factors) explain the disconnection between tau pathology and brain atrophy (brain resilience), and tau pathology and cognitive decline (cognitive resilience). METHODS We included 428 amyloid positive participants (134 cognitively unimpaired (CU), 128 with mild cognitive impairment (MCI), 166 with AD dementia) from the BioFINDER-2 study. At baseline, participants underwent tau positron emission tomography (tau-PET), magnetic resonance imaging (MRI), cognitive testing, and lumbar puncture. Longitudinal data were available for MRI (mean (standard deviation) follow-up 26.4 (10.7) months) and cognition (25.2 (11.4) months). We analysed 18 pre-selected CSF proteins, reflecting vascular, synaptic, and axonal integrity, neuroinflammation, and neurotrophic factors. Stratifying by cognitive status, we performed linear mixed-effects models with cortical thickness (brain resilience) and global cognition (cognitive resilience) as dependent variables to assess whether the CSF biomarkers interacted with tau-PET levels in its effect on cortical atrophy and cognitive decline. RESULTS Regarding brain resilience, interaction effects were observed in AD dementia, with vascular integrity biomarkers (VEGF-A (βinteraction = -0.009, pFDR = 0.047) and VEGF-B (βinteraction = -0.010, pFDR = 0.037)) negatively moderating the association between tau-PET signal and atrophy. In MCI, higher NfL levels were associated with more longitudinal cortical atrophy (β = -0.109, pFDR = 0.033) and lower baseline cortical thickness (β = -0.708, pFDR = 0.033) controlling for tau-PET signal. Cognitive resilience analyses in CU revealed interactions with tau-PET signal for inflammatory (GFAP, IL-15; βinteraction -0.073--0.069, pFDR 0.001-0.045), vascular (VEGF-A, VEGF-D, PGF; βinteraction -0.099--0.063, pFDR < 0.001-0.046), synaptic (14-3-3ζ/δ; βinteraction = -0.092, pFDR = 0.041), axonal (NfL; βinteraction = -0.079, pFDR < 0.001), and neurotrophic (NGF; βinteraction = 0.091, pFDR < 0.001) biomarkers. In MCI higher NfL levels (βmain = -0.690, pFDR = 0.025) were associated with faster cognitive decline independent of tau-PET signal. CONCLUSIONS Biomarkers of co-existing pathological processes, in particular vascular pathology and axonal degeneration, interact with levels of tau pathology on its association with the downstream effects of AD pathology (i.e. brain atrophy and cognitive decline). This indicates that vascular pathology and axonal degeneration could impact brain and cognitive resilience.
Collapse
Affiliation(s)
- Anna L Svenningsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden.
| | - Diana I Bocancea
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081, Amsterdam, The Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Anita van Loenhoud
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV, Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Center for Medical Image Computing, University College London, London, WC1N 3BG, UK
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Department of Neurology, Skåne University Hospital, 211 84, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Daniilidou M, Holleman J, Hagman G, Kåreholt I, Aspö M, Brinkmalm A, Zetterberg H, Blennow K, Solomon A, Kivipelto M, Sindi S, Matton A. Neuroinflammation, cerebrovascular dysfunction and diurnal cortisol biomarkers in a memory clinic cohort: Findings from the Co-STAR study. Transl Psychiatry 2024; 14:364. [PMID: 39251589 PMCID: PMC11385239 DOI: 10.1038/s41398-024-03072-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024] Open
Abstract
Cortisol dysregulation, neuroinflammation, and cerebrovascular dysfunction are biological processes that have been separately shown to be affected in Alzheimer's disease (AD). Here, we aimed to identify biomarker signatures reflecting these pathways in 108 memory clinic patients with subjective cognitive decline (SCD, N = 40), mild cognitive impairment (MCI, N = 39), and AD (N = 29). Participants were from the well-characterized Cortisol and Stress in Alzheimer's Disease (Co-STAR) cohort, recruited at Karolinska University Hospital. Salivary diurnal cortisol measures and 41 CSF proteins were analyzed. Principal component analysis was applied to identify combined biosignatures related to AD pathology, synaptic loss, and neuropsychological assessments, in linear regressions adjusted for confounders, such as age, sex, education and diagnosis. We found increased CSF levels of C-reactive protein (CRP), interferon γ-inducible protein (IP-10), thymus and activation-regulated chemokine (TARC), intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) in MCI patients. Further, markers of cortisol dysregulation (flattened salivary cortisol awakening response and flattened cortisol slope) correlated with increased levels of placental growth factor (PlGF), IP-10, and chitinase 3-like 1 (YKL-40) in the total cohort. A biosignature composed of cortisol awakening response, cortisol slope, and CSF IL-6 was downregulated in AD patients. Moreover, biomarker signatures reflecting overlapping pathophysiological processes of neuroinflammation and vascular injury were associated with AD pathology, synaptic loss, and worsened processing speed. Our findings suggest an early dysregulation of immune and cerebrovascular processes during the MCI stage and provide insights into the interrelationship of chronic stress and neuroinflammation in AD.
Collapse
Affiliation(s)
- Makrina Daniilidou
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Jasper Holleman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Göran Hagman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Ingemar Kåreholt
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Institute of Gerontology, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Malin Aspö
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Alina Solomon
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Shireen Sindi
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Anna Matton
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
17
|
Debette S, Caro I, Western D, Namba S, Sun N, Kawaguchi S, He Y, Fujita M, Roshchupkin G, D'Aoust T, Duperron MG, Sargurupremraj M, Tsuchida A, Koido M, Ahmadi M, Yang C, Timsina J, Ibanez L, Matsuda K, Suzuki Y, Oda Y, Kanai A, Jandaghi P, Munter HM, Auld D, Astafeva I, Puerta R, Rotter J, Psaty B, Bis J, Longstreth W, Couffinhal T, Garcia-Gonzalez P, Pytel V, Marquié M, Cano A, Boada M, Joliot M, Lathrop M, Le Grand Q, Launer L, Wardlaw J, Heiman M, Ruiz A, Matthews P, Seshadri S, Fornage M, Adams H, Mishra A, Trégouët DA, Okada Y, Kellis M, De Jager P, Tzourio C, Kamatani Y, Matsuda F, Cruchaga C. Proteogenomics in cerebrospinal fluid and plasma reveals new biological fingerprint of cerebral small vessel disease. RESEARCH SQUARE 2024:rs.3.rs-4535534. [PMID: 39011113 PMCID: PMC11247936 DOI: 10.21203/rs.3.rs-4535534/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Cerebral small vessel disease (cSVD) is a leading cause of stroke and dementia with no specific mechanism-based treatment. We used Mendelian randomization to combine a unique cerebrospinal fluid (CSF) and plasma pQTL resource with the latest European-ancestry GWAS of MRI-markers of cSVD (white matter hyperintensities, perivascular spaces). We describe a new biological fingerprint of 49 protein-cSVD associations, predominantly in the CSF. We implemented a multipronged follow-up, across fluids, platforms, and ancestries (Europeans and East-Asian), including testing associations of direct plasma protein measurements with MRI-cSVD. We highlight 16 proteins robustly associated in both CSF and plasma, with 24/4 proteins identified in CSF/plasma only. cSVD-proteins were enriched in extracellular matrix and immune response pathways, and in genes enriched in microglia and specific microglial states (integration with single-nucleus RNA sequencing). Immune-related proteins were associated with MRI-cSVD already at age twenty. Half of cSVD-proteins were associated with stroke, dementia, or both, and seven cSVD-proteins are targets for known drugs (used for other indications in directions compatible with beneficial therapeutic effects. This first cSVD proteogenomic signature opens new avenues for biomarker and therapeutic developments.
Collapse
Affiliation(s)
| | | | - Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Na Sun
- MIT Computer Science and Artificial Intelligence Laboratory; Broad Institute of MIT and Harvard
| | | | - Yunye He
- Graduate School of Frontier Sciences, The University of Tokyo
| | | | | | - Tim D'Aoust
- Bordeaux Population Health, Inserm U1219, University of Bordeaux
| | | | - Murali Sargurupremraj
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000 Bordeaux, France; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases Unive
| | | | - Masaru Koido
- Graduate School of Frontier Sciences, The University of Tokyo
| | | | | | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Koichi Matsuda
- Department of Computational Biology and Medical Sciences, Graduate school of Frontier Sciences, The University of Tokyo
| | | | - Yoshiya Oda
- Graduate School of Medicine, The University of Tokyo
| | | | | | | | - Dan Auld
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University
| | - Iana Astafeva
- Bordeaux Population Health, Inserm U1219, University of Bordeaux; Institute of Neurodegenerative Diseases
| | | | - Jerome Rotter
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center
| | | | | | | | - Thierry Couffinhal
- University of Bordeaux, The clinical unit of Exploration, Prevention and Care Center for Atherosclerosis (CEPTA), CHUB, Inserm U1034
| | | | - Vanesa Pytel
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya; CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III
| | | | | | | | | | - Mark Lathrop
- Department of Human Genetics, McGill University, 1205 Dr Penfield Avenue, Montreal, QC, H3A 1B1, Canada
| | - Quentin Le Grand
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219
| | - Lenore Launer
- National Institute on Aging, National Institutes of Health
| | | | | | - Agustin Ruiz
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center; Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya;CIBERN
| | - Paul Matthews
- UK Dementia Research Institute Centre at Imperial College London
| | | | - Myriam Fornage
- 1. Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center 2. Human Genetics Center, Department of Epidemiology, School of Public Health
| | - Hieab Adams
- Department of Human Genetics, Radboud University Medical Center; Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez
| | | | | | - Yukinori Okada
- Department of Genome Informatics, Graduate School of Medicine, The Univ. of Tokyo; Department of Statistical Genetics, Osaka Univ. Graduate School of Medicine; Laboratory for Systems Genetic, RIKEN
| | | | | | | | | | | | | |
Collapse
|
18
|
Lu J, Wang X, Xu F, Rao C, Guo Y, Su Z, Chen S, Li Q. Exploring causal correlations of inflammatory biomarkers in idiopathic normal-pressure hydrocephalus: insights from bidirectional Mendelian randomization analysis. Front Aging Neurosci 2024; 16:1412434. [PMID: 38974901 PMCID: PMC11224557 DOI: 10.3389/fnagi.2024.1412434] [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: 04/04/2024] [Accepted: 06/11/2024] [Indexed: 07/09/2024] Open
Abstract
Background and objective Neuroinflammatory processes have been identified as playing a crucial role in the pathophysiology of various neurodegenerative diseases, including idiopathic normal-pressure hydrocephalus (iNPH). iNPH, defined as a common disease of cognitive impairment in older adults, poses major challenges for therapeutic interventions owing to the stringent methodological requirements of relevant studies, clinical heterogeneity, unclear etiology, and uncertain diagnostic criteria. This study aims to assess the relationship between circulating inflammatory biomarkers and iNPH risk using bidirectional two-sample Mendelian randomization (MR) combined with meta-analysis. Methods In our bidirectional MR study, genetic data from a genome-wide association study (GWAS) involving 1,456 iNPH cases and 409,726 controls of European ancestry were employed. Single-nucleotide polymorphisms (SNPs) associated with exposures served as instrumental variables for estimating the causal relationships between iNPH and 132 types of circulating inflammatory biomarkers from corresponding GWAS data. Causal associations were primarily examined using the inverse variance-weighted method, supplemented by MR-Egger, weighted median, simple mode, and weighted mode analyses. In the results, heterogeneity was assessed using the Cochran Q test. Horizontal pleiotropy was evaluated through the MR-Egger intercept test and the MR pleiotropy residual sum and outliers test. Sensitivity analysis was conducted through leave-one-out analysis. Reverse MR analyses were performed to mitigate bias from reverse causality. Meta-analyses of identical inflammatory biomarkers from both data sources strengthened the findings. Results Results indicated a genetically predicted association between Interleukin-16 (IL-16) [OR: 1.228, 95% CI: 1.049-1.439, p = 0.011], TNF-related apoptosis ligand (TRAIL) [OR: 1.111, 95% CI: 1.019-1.210, p = 0.017] and Urokinase-type plasminogen activator (uPA) [OR: 1.303, 95% CI: 1.025-1.658, p = 0.031] and the risk of iNPH. Additionally, changes in human Glial cell line-derived neurotrophic factor (hGDNF) [OR: 1.044, 95% CI: 1.006-1.084, p = 0.023], Matrix metalloproteinase-1 (MMP-1) [OR: 1.058, 95% CI: 1.020, 1.098, p = 0.003] and Interleukin-12p70 (IL-12p70) [OR: 0.897, 95% CI: 0.946-0.997, p = 0.037] levels were identified as possible consequences of iNPH. Conclusion Our MR study of inflammatory biomarkers and iNPH, indicated that IL-16, TRAIL, and uPA contribute to iNPH pathogenesis. Furthermore, iNPH may influence the expression of hGDNF, MMP-1, and IL-12p70. Therefore, targeting specific inflammatory biomarkers could be promising strategy for future iNPH treatment and prevention.
Collapse
Affiliation(s)
- Jianglong Lu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xianpeng Wang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fanjie Xu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Changjun Rao
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuhang Guo
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhipeng Su
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Siyan Chen
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Li
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
19
|
Yang HS, Yau WYW, Carlyle BC, Trombetta BA, Zhang C, Shirzadi Z, Schultz AP, Pruzin JJ, Fitzpatrick CD, Kirn DR, Rabin JS, Buckley RF, Hohman TJ, Rentz DM, Tanzi RE, Johnson KA, Sperling RA, Arnold SE, Chhatwal JP. Plasma VEGFA and PGF impact longitudinal tau and cognition in preclinical Alzheimer's disease. Brain 2024; 147:2158-2168. [PMID: 38315899 PMCID: PMC11146430 DOI: 10.1093/brain/awae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/08/2023] [Accepted: 12/21/2023] [Indexed: 02/07/2024] Open
Abstract
Vascular dysfunction is increasingly recognized as an important contributor to the pathogenesis of Alzheimer's disease. Alterations in vascular endothelial growth factor (VEGF) pathways have been implicated as potential mechanisms. However, the specific impact of VEGF proteins in preclinical Alzheimer's disease and their relationships with other Alzheimer's disease and vascular pathologies during this critical early period remain to be elucidated. We included 317 older adults from the Harvard Aging Brain Study, a cohort of individuals who were cognitively unimpaired at baseline and followed longitudinally for up to 12 years. Baseline VEGF family protein levels (VEGFA, VEGFC, VEGFD, PGF and FLT1) were measured in fasting plasma using high-sensitivity immunoassays. Using linear mixed effects models, we examined the interactive effects of baseline plasma VEGF proteins and amyloid PET burden (Pittsburgh Compound-B) on longitudinal cognition (Preclinical Alzheimer Cognitive Composite-5). We further investigated if effects on cognition were mediated by early neocortical tau accumulation (flortaucipir PET burden in the inferior temporal cortex) or hippocampal atrophy. Lastly, we examined the impact of adjusting for baseline cardiovascular risk score or white matter hyperintensity volume. Baseline plasma VEGFA and PGF each showed a significant interaction with amyloid burden on prospective cognitive decline. Specifically, low VEGFA and high PGF were associated with greater cognitive decline in individuals with elevated amyloid, i.e. those on the Alzheimer's disease continuum. Concordantly, low VEGFA and high PGF were associated with accelerated longitudinal tau accumulation in those with elevated amyloid. Moderated mediation analyses confirmed that accelerated tau accumulation fully mediated the effects of low VEGFA and partially mediated (31%) the effects of high PGF on faster amyloid-related cognitive decline. The effects of VEGFA and PGF on tau and cognition remained significant after adjusting for cardiovascular risk score or white matter hyperintensity volume. There were concordant but non-significant associations with longitudinal hippocampal atrophy. Together, our findings implicate low VEGFA and high PGF in accelerating early neocortical tau pathology and cognitive decline in preclinical Alzheimer's disease. Additionally, our results underscore the potential of these minimally-invasive plasma biomarkers to inform the risk of Alzheimer's disease progression in the preclinical population. Importantly, VEGFA and PGF appear to capture distinct effects from vascular risks and cerebrovascular injury. This highlights their potential as new therapeutic targets, in combination with anti-amyloid and traditional vascular risk reduction therapies, to slow the trajectory of preclinical Alzheimer's disease and delay or prevent the onset of cognitive decline.
Collapse
Affiliation(s)
- Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Wai-Ying Wendy Yau
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Becky C Carlyle
- Harvard Medical School, Boston, MA 02115, USA
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Physiology, Anatomy and Genetics, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3PT, UK
| | - Bianca A Trombetta
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Can Zhang
- Harvard Medical School, Boston, MA 02115, USA
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Zahra Shirzadi
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Jeremy J Pruzin
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Neurology, Banner Alzheimer’s Institute, Phoenix, AZ 85006, USA
| | | | - Dylan R Kirn
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Jennifer S Rabin
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Department of Medicine, Rehabilitation Sciences Institute, University of Toronto, Toronto, ON M5G 1V7, Canada
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Rudolph E Tanzi
- Harvard Medical School, Boston, MA 02115, USA
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Steven E Arnold
- Harvard Medical School, Boston, MA 02115, USA
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
20
|
Hayashi H, Sone T, Iokawa K, Sumigawa K, Fujita T, Kawamata H, Asao A, Kawasaki I, Ogasawara M, Kawakatsu S. Effects of computerized cognitive training on biomarker responses in older adults with mild cognitive impairment: A scoping review. Health Sci Rep 2024; 7:e2175. [PMID: 38895550 PMCID: PMC11182779 DOI: 10.1002/hsr2.2175] [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: 09/29/2023] [Revised: 03/08/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Background and Aims Mild cognitive impairment (MCI) is a widespread condition in older individuals, posing significant risk of dementia. However, limited research has been conducted to explore effective interventions and clarify their impact at the neural level. Therefore, this study aimed to investigate the effects of computerized cognitive training (CCT) and explore the associated neural mechanisms in preventing dementia in older individuals with MCI, with a view to inform future intervention efforts. Methods We reviewed the effects of CCT on biomarker outcomes in older adults with MCI. The search was conducted for studies published between 2010 and May 10, 2023, using three search engines: PubMed, Scopus, and Cumulative Index to Nursing and Allied Health Literature. The inclusion criteria were as follows: studies that involved participants diagnosed with MCI, included CCT, included quantitative assessment of biomarker results, and conducted randomized controlled trials. Results Sixteen studies that used biomarkers, including magnetic resonance imaging, electroencephalography (EEG), functional near-infrared spectroscopy (fNIRS), and blood or salivary biomarkers, were extracted. The results showed that CCT caused changes in structure and function within the main brain network, including the default mode network, and decreased both theta rhythm activity on EEG and prefrontal activity on fNIRS, with improvement in cognitive function. Furthermore, CCT combined with physical exercise showed more significant structural and functional changes in extensive brain regions compared with CCT alone. Virtual reality-based cognitive training improved not only executive function but also instrumental activities of daily living. Conclusion CCT causes functional and structural changes in extensive brain regions and improves cognitive function in older adults with MCI. Our findings highlight the potential of individualized intervention methods and biomarker assessment according to the specific causes of MCI. Future research should aim to optimize these personalized therapeutic strategies to maximize the benefits of CCT in older adults with MCI.
Collapse
Affiliation(s)
- Hiroshi Hayashi
- Department of Occupational TherapyFukushima Medical University School of Health SciencesFukushimaJapan
| | - Toshimasa Sone
- Department of Occupational TherapyFukushima Medical University School of Health SciencesFukushimaJapan
| | - Kazuaki Iokawa
- Department of Occupational TherapyFukushima Medical University School of Health SciencesFukushimaJapan
| | - Koshi Sumigawa
- Department of Occupational TherapyFukushima Medical University School of Health SciencesFukushimaJapan
| | - Takaaki Fujita
- Department of Occupational TherapyFukushima Medical University School of Health SciencesFukushimaJapan
| | - Hironori Kawamata
- Department of Occupational TherapyFukushima Medical University School of Health SciencesFukushimaJapan
| | - Akihiko Asao
- Department of Occupational TherapyFukushima Medical University School of Health SciencesFukushimaJapan
| | - Iori Kawasaki
- Department of Occupational TherapyFukushima Medical University School of Health SciencesFukushimaJapan
| | - Maki Ogasawara
- Department of Occupational TherapyFukushima Medical University School of Health SciencesFukushimaJapan
| | - Shinobu Kawakatsu
- Department of Neuropsychiatry, Aizu Medical CenterFukushima Medical UniversityAizuwakamatsuJapan
| |
Collapse
|
21
|
Lim YA, Tan LS, Lee WT, Sim WL, Lv Y, Takakuni M, Saito S, Ihara M, Arumugam TV, Chen C, Wong FWS, Dawe GS. Hope for vascular cognitive impairment: Ac-YVAD-cmk as a novel treatment against white matter rarefaction. PLoS One 2024; 19:e0299703. [PMID: 38630707 PMCID: PMC11023579 DOI: 10.1371/journal.pone.0299703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/14/2024] [Indexed: 04/19/2024] Open
Abstract
Vascular cognitive impairment (VCI) is the second leading cause of dementia with limited treatment options, characterised by cerebral hypoperfusion-induced white matter rarefaction (WMR). Subcortical VCI is the most common form of VCI, but the underlying reasons for region susceptibility remain elusive. Recent studies employing the bilateral cortical artery stenosis (BCAS) method demonstrate that various inflammasomes regulate white matter injury and blood-brain barrier dysfunction but whether caspase-1 inhibition will be beneficial remains unclear. To address this, we performed BCAS on C57/BL6 mice to study the effects of Ac-YVAD-cmk, a caspase-1 inhibitor, on the subcortical and cortical regions. Cerebral blood flow (CBF), WMR, neuroinflammation and the expression of tight junction-related proteins associated with blood-brain barrier integrity were assessed 15 days post BCAS. We observed that Ac-YVAD-cmk restored CBF, attenuated BCAS-induced WMR and restored subcortical myelin expression. Within the subcortical region, BCAS activated the NLRP3/caspase-1/interleukin-1beta axis only within the subcortical region, which was attenuated by Ac-YVAD-cmk. Although we observed that BCAS induced significant increases in VCAM-1 expression in both brain regions that were attenuated with Ac-YVAD-cmk, only ZO-1 and occludin were observed to be significantly altered in the subcortical region. Here we show that caspase-1 may contribute to subcortical regional susceptibility in a mouse model of VCI. In addition, our results support further investigations into the potential of Ac-YVAD-cmk as a novel treatment strategy against subcortical VCI and other conditions exhibiting cerebral hypoperfusion-induced WMR.
Collapse
Affiliation(s)
- Yun-An Lim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Si Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Thye Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Liang Sim
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Yang Lv
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Maki Takakuni
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
- Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | | | - Christopher Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Fred Wai-Shiu Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gavin Stewart Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) is an all-encompassing term that describes cognitive impairment due to cerebrovascular origins. With the advancement of imaging and pathological studies, we now understand that VCID is often comorbid with Alzheimer disease. While researchers in the Alzheimer disease field have been working for years to establish and test blood-based biomarkers for Alzheimer disease diagnosis, prognosis, clinical therapy discovery, and early detection, blood-based biomarkers for VCID are in their infancy and also face challenges. VCID is heterogeneous, comprising many different pathological entities (ischemic, or hemorrhagic), and spatial and temporal differences (acute or chronic). This review highlights pathways that are aiding the search for sensitive and specific blood-based cerebrovascular dysfunction markers, describes promising candidates, and explains ongoing initiatives to discover blood-based VCID biomarkers.
Collapse
Affiliation(s)
- Kate E. Foley
- Stark Neurosciences Research Institute, Indiana University, Indianapolis IN, USA
- Department of Neurology, School of Medicine, Indiana University, Indianapolis IN, USA
| | - Donna M. Wilcock
- Stark Neurosciences Research Institute, Indiana University, Indianapolis IN, USA
- Department of Neurology, School of Medicine, Indiana University, Indianapolis IN, USA
| |
Collapse
|
23
|
Karvelas N, Oh B, Wang E, Cobigo Y, Tsuei T, Fitzsimons S, Younes K, Ehrenberg A, Geschwind MD, Schwartz D, Kramer JH, Ferguson AR, Miller BL, Silbert LC, Rosen HJ, Elahi FM. Enlarged perivascular spaces are associated with white matter injury, cognition and inflammation in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Brain Commun 2024; 6:fcae071. [PMID: 38495305 PMCID: PMC10943571 DOI: 10.1093/braincomms/fcae071] [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: 10/09/2023] [Revised: 01/18/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024] Open
Abstract
Enlarged perivascular spaces have been previously reported in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, but their significance and pathophysiology remains unclear. We investigated associations of white matter enlarged perivascular spaces with classical imaging measures, cognitive measures and plasma proteins to better understand what enlarged perivascular spaces represent in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy and whether radiographic measures of enlarged perivascular spaces would be of value in future therapeutic discovery studies for cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Twenty-four individuals with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy and 24 age- and sex-matched controls were included. Disease status was determined based on the presence of NOTCH3 mutation. Brain imaging measures of white matter hyperintensity, brain parenchymal fraction, white matter enlarged perivascular space volumes, clinical and cognitive measures as well as plasma proteomics were used in models. White matter enlarged perivascular space volumes were calculated via a novel, semiautomated pipeline, and levels of 7363 proteins were quantified in plasma using the SomaScan assay. The relationship of enlarged perivascular spaces with global burden of white matter hyperintensity, brain atrophy, functional status, neurocognitive measures and plasma proteins was modelled with linear regression models. Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy and control groups did not exhibit differences in mean enlarged perivascular space volumes. However, increased enlarged perivascular space volumes in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy were associated with increased white matter hyperintensity volume (β = 0.57, P = 0.05), Clinical Dementia Rating Sum-of-Boxes score (β = 0.49, P = 0.04) and marginally with decreased brain parenchymal fraction (β = -0.03, P = 0.10). In interaction term models, the interaction term between cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy disease status and enlarged perivascular space volume was associated with increased white matter hyperintensity volume (β = 0.57, P = 0.02), Clinical Dementia Rating Sum-of-Boxes score (β = 0.52, P = 0.02), Mini-Mental State Examination score (β = -1.49, P = 0.03) and marginally with decreased brain parenchymal fraction (β = -0.03, P = 0.07). Proteins positively associated with enlarged perivascular space volumes were found to be related to leukocyte migration and inflammation, while negatively associated proteins were related to lipid metabolism. Two central hub proteins were identified in protein networks associated with enlarged perivascular space volumes: CXC motif chemokine ligand 8/interleukin-8 and C-C motif chemokine ligand 2/monocyte chemoattractant protein 1. The levels of CXC motif chemokine ligand 8/interleukin-8 were also associated with increased white matter hyperintensity volume (β = 42.86, P = 0.03), and levels of C-C motif chemokine ligand 2/monocyte chemoattractant protein 1 were further associated with decreased brain parenchymal fraction (β = -0.0007, P < 0.01) and Mini-Mental State Examination score (β = -0.02, P < 0.01) and increased Trail Making Test B completion time (β = 0.76, P < 0.01). No proteins were associated with all three studied imaging measures of pathology (brain parenchymal fraction, enlarged perivascular spaces, white matter hyperintensity). Based on associations uncovered between enlarged perivascular space volumes and cognitive functions, imaging and plasma proteins, we conclude that white matter enlarged perivascular space volumes may capture pathologies contributing to chronic brain dysfunction and degeneration in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy.
Collapse
Affiliation(s)
- Nikolaos Karvelas
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bradley Oh
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Earnest Wang
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Yann Cobigo
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Torie Tsuei
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Stephen Fitzsimons
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kyan Younes
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94304, USA
| | - Alexander Ehrenberg
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael D Geschwind
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Daniel Schwartz
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Adam R Ferguson
- Department of Neurological surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94110, USA
- San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Lisa C Silbert
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
- NIA-Layton Alzheimer’s Disease Research Center, Oregon Health & Science University, Portland, OR 97239, USA
- Portland Veterans Affairs Health Care System, Portland, OR 97239, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Fanny M Elahi
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| |
Collapse
|
24
|
der Heijden HV, Rameh V, Golden E, Ronen I, Sundel RP, Knight A, Chang JC, Upadhyay J. Implications of Inflammatory Processes on a Developing Central Nervous System in Childhood-Onset Systemic Lupus Erythematosus. Arthritis Rheumatol 2024; 76:332-344. [PMID: 37901986 PMCID: PMC10922196 DOI: 10.1002/art.42736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/20/2023] [Accepted: 10/25/2023] [Indexed: 10/31/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that is increasingly affecting pediatric and adult populations. Neuropsychiatric manifestations (ie, cognitive dysfunction and mood disorders) appear to occur with greater severity and poorer prognosis in childhood-onset SLE (cSLE) versus adult-onset SLE, negatively impacting school function, self-management, and psychosocial health, as well as lifelong health-related quality of life. In this review, we describe pathogenic mechanisms active in cSLE, such as maladaptive inflammatory processes and ischemia, which are hypothesized to underpin central phenotypes in patients with cSLE, and the role of alterations in protective central nervous system (CNS) barriers (ie, the blood-brain barrier) are also discussed. Recent findings derived from novel neuroimaging approaches are highlighted because the methods employed in these studies hold potential for identifying CNS abnormalities that would otherwise remain undetected with conventional multiple resonance imaging studies (eg, T2-weighted or fluid-attenuated inversion recovery sequences). Furthermore, we propose that a more robust presentation of neuropsychiatric symptoms in cSLE is in part due to the harmful impact of a chronic inflammatory insult on a developing CNS. Although the immature status of the CNS may leave patients with cSLE more vulnerable to harboring neuropsychiatric manifestations, the same property may represent a greater urgency to reverse the maladaptive effects associated with a proneuroinflammatory state, provided that effective diagnostic tools and treatment strategies are available. Finally, considering the crosstalk among the CNS and other organ systems affected in cSLE, we postulate that a finer understanding of this interconnectivity and its role in the clinical presentation in cSLE is warranted.
Collapse
Affiliation(s)
- Hanne Van der Heijden
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
- Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Vanessa Rameh
- Division of Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Emma Golden
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Itamar Ronen
- Clinical Imaging Science Center, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Robert P. Sundel
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Andrea Knight
- Division of Rheumatology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
- Neurosciences and Mental Health, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Joyce C. Chang
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Jaymin Upadhyay
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA USA
| |
Collapse
|
25
|
van Amerongen S, Pulukuri SV, Tuz-Zahra F, Tripodis Y, Cherry JD, Bernick C, Geda YE, Wethe JV, Katz DI, Alosco ML, Adler CH, Balcer LJ, Ashton NJ, Blennow K, Zetterberg H, Daneshvar DH, Colasurdo EA, Iliff JJ, Li G, Peskind ER, Shenton ME, Reiman EM, Cummings JL, Stern RA. Inflammatory biomarkers for neurobehavioral dysregulation in former American football players: findings from the DIAGNOSE CTE Research Project. J Neuroinflammation 2024; 21:46. [PMID: 38336728 PMCID: PMC10854026 DOI: 10.1186/s12974-024-03034-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Traumatic encephalopathy syndrome (TES) is defined as the clinical manifestation of the neuropathological entity chronic traumatic encephalopathy (CTE). A core feature of TES is neurobehavioral dysregulation (NBD), a neuropsychiatric syndrome in repetitive head impact (RHI)-exposed individuals, characterized by a poor regulation of emotions/behavior. To discover biological correlates for NBD, we investigated the association between biomarkers of inflammation (interleukin (IL)-1β, IL-6, IL-8, IL-10, C-reactive protein (CRP), tumor necrosis factor (TNF)-α) in cerebrospinal fluid (CSF) and NBD symptoms in former American football players and unexposed individuals. METHODS Our cohort consisted of former American football players, with (n = 104) or without (n = 76) NBD diagnosis, as well as asymptomatic unexposed individuals (n = 55) from the DIAGNOSE CTE Research Project. Specific measures for NBD were derived (i.e., explosivity, emotional dyscontrol, impulsivity, affective lability, and a total NBD score) from a factor analysis of multiple self-report neuropsychiatric measures. Analyses of covariance tested differences in biomarker concentrations between the three groups. Within former football players, multivariable linear regression models assessed relationships among log-transformed inflammatory biomarkers, proxies for RHI exposure (total years of football, cumulative head impact index), and NBD factor scores, adjusted for relevant confounding variables. Sensitivity analyses tested (1) differences in age subgroups (< 60, ≥ 60 years); (2) whether associations could be identified with plasma inflammatory biomarkers; (3) associations between neurodegeneration and NBD, using plasma neurofilament light (NfL) chain protein; and (4) associations between biomarkers and cognitive performance to explore broader clinical symptoms related to TES. RESULTS CSF IL-6 was higher in former American football players with NBD diagnosis compared to players without NBD. Furthermore, elevated levels of CSF IL-6 were significantly associated with higher emotional dyscontrol, affective lability, impulsivity, and total NBD scores. In older football players, plasma NfL was associated with higher emotional dyscontrol and impulsivity, but also with worse executive function and processing speed. Proxies for RHI exposure were not significantly associated with biomarker concentrations. CONCLUSION Specific NBD symptoms in former American football players may result from multiple factors, including neuroinflammation and neurodegeneration. Future studies need to unravel the exact link between NBD and RHI exposure, including the role of other pathophysiological pathways.
Collapse
Affiliation(s)
- Suzan van Amerongen
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Surya V Pulukuri
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Fatima Tuz-Zahra
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yorghos Tripodis
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jonathan D Cherry
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Charles Bernick
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Yonas E Geda
- Department of Neurology and the Franke Global Neuroscience Education Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Jennifer V Wethe
- Department of Psychiatry and Psychology, Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Douglas I Katz
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Brain Injury Program, Encompass Health Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Michael L Alosco
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Laura J Balcer
- Departments of Neurology, Population Health and Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Daniel H Daneshvar
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Colasurdo
- Veterans Affairs Northwest Mental Illness Research, Education, and Clinical Center, Seattle, WA, USA
| | - Jeffrey J Iliff
- Veterans Affairs Northwest Mental Illness Research, Education, and Clinical Center, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Gail Li
- Veterans Affairs Northwest Mental Illness Research, Education, and Clinical Center, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System Geriatric Research, Seattle, WA, USA
| | - Elaine R Peskind
- Veterans Affairs Northwest Mental Illness Research, Education, and Clinical Center, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Martha E Shenton
- Psychiatry Neuroimaging Laboratory, Harvard Medical School, Departments of Psychiatry and Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Eric M Reiman
- Banner Alzheimer's Institute, University of Arizona, Arizona State University, Translational Genomics Research Institute, and Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Robert A Stern
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Departments of Neurosurgery, and Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
26
|
Wu LY, Chong JR, Chong JPC, Hilal S, Venketasubramanian N, Tan BY, Richards AM, Chen CP, Lai MKP. Serum Placental Growth Factor as a Marker of Cerebrovascular Disease Burden in Alzheimer's Disease. J Alzheimers Dis 2024; 97:1289-1298. [PMID: 38217598 DOI: 10.3233/jad-230811] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
BACKGROUND Concomitant cerebrovascular diseases (CeVD) have been identified as an important determinant of Alzheimer's disease (AD) progression. Development of robust blood-based biomarkers will provide critical tools to evaluate prognosis and potential interventional strategies for AD with CeVD. OBJECTIVE This study investigated circulating placental growth factor (PlGF), a potent pro-angiogenic factor related to endothelial dysfunction and vascular inflammation, in an Asian memory clinic cohort of non-demented individuals as well as AD, including its associations with neuroimaging markers of CeVD. METHODS 109 patients with AD, 76 cognitively impaired with no dementia (CIND), and 56 non-cognitively impaired (NCI) were included in this cross-sectional study. All subjects underwent 3T brain magnetic resonance imaging to assess white matter hyperintensities (WMH), lacunes, cortical infarcts, and cerebral microbleeds (CMBs). Serum PlGF concentrations were measured by electrochemiluminescence immunoassays. RESULTS Serum PlGF was elevated in AD, but not CIND, compared to the NCI controls. Adjusted concentrations of PlGF were associated with AD only in the presence of significant CeVD. Elevated PlGF was significantly associated with higher burden of WMH and with CMBs in AD patients. CONCLUSIONS Serum PlGF has potential utility as a biomarker for the presence of CeVD, specifically WMH and CMBs, in AD. Further studies are needed to elucidate the underlying pathophysiological mechanisms linking PlGF to CeVD, as well as to further assess PlGF's clinical utility.
Collapse
Affiliation(s)
- Liu-Yun Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Jenny P C Chong
- Cardiovascular Research Institute, National University Heart Centre, Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | | | | | - Arthur Mark Richards
- Cardiovascular Research Institute, National University Heart Centre, Singapore
- Department of Medicine, National University Health System, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| |
Collapse
|
27
|
Jaime Garcia D, Chagnot A, Wardlaw JM, Montagne A. A Scoping Review on Biomarkers of Endothelial Dysfunction in Small Vessel Disease: Molecular Insights from Human Studies. Int J Mol Sci 2023; 24:13114. [PMID: 37685924 PMCID: PMC10488088 DOI: 10.3390/ijms241713114] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Small vessel disease (SVD) is a highly prevalent disorder of the brain's microvessels and a common cause of dementia as well as ischaemic and haemorrhagic strokes. Though much about the underlying pathophysiology of SVD remains poorly understood, a wealth of recently published evidence strongly suggests a key role of microvessel endothelial dysfunction and a compromised blood-brain barrier (BBB) in the development and progression of the disease. Understanding the causes and downstream consequences associated with endothelial dysfunction in this pathological context could aid in the development of effective diagnostic and prognostic tools and provide promising avenues for potential therapeutic interventions. In this scoping review, we aim to summarise the findings from clinical studies examining the role of the molecular mechanisms underlying endothelial dysfunction in SVD, focussing on biochemical markers of endothelial dysfunction detectable in biofluids, including cell adhesion molecules, BBB transporters, cytokines/chemokines, inflammatory markers, coagulation factors, growth factors, and markers involved in the nitric oxide cascade.
Collapse
Affiliation(s)
- Daniela Jaime Garcia
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; (D.J.G.); (J.M.W.)
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK;
| | - Audrey Chagnot
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK;
| | - Joanna M. Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; (D.J.G.); (J.M.W.)
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK;
| | - Axel Montagne
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; (D.J.G.); (J.M.W.)
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK;
| |
Collapse
|
28
|
Schmidt-Morgenroth I, Michaud P, Gasparini F, Avrameas A. Central and Peripheral Inflammation in Mild Cognitive Impairment in the Context of Alzheimer's Disease. Int J Mol Sci 2023; 24:10523. [PMID: 37445700 DOI: 10.3390/ijms241310523] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/05/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Mild cognitive impairment (MCI) is characterized by an abnormal decline in mental and cognitive function compared with normal cognitive aging. It is an underlying condition of Alzheimer's disease (AD), an irreversible neurodegenerative disease. In recent years, neuroinflammation has been investigated as a new leading target that contributes to MCI progression into AD. Understanding the mechanism underlying inflammatory processes involved in the early onset of the disease could help find a safe and effective way to diagnose and treat patients. In this article, we assessed over twenty different blood and cerebrospinal fluid (CSF) inflammatory biomarker concentrations with immunoassay methods in patients with MCI (mild cognitive impairment), non-impaired control (NIC), and serum healthy control (HC). We performed group comparisons and analyzed in-group correlations between the biomarkers. We included 107 participants (mean age: 64.7 ± 7.8, women: 58.9%). CSF osteopontin and YKL-40 were significantly increased in the MCI group, whereas serum C-reactive protein and interleukin-6 were significantly higher (p < 0.001) in the NIC group compared with the MCI and HC groups. Stronger correlations between interleukin-1β and inflammasome markers were observed in the serum of the MCI group. We confirmed specific inflammatory activation in the central nervous system and interleukin-1β pathway upregulation in the serum of the MCI cohort.
Collapse
Affiliation(s)
- Inès Schmidt-Morgenroth
- Novartis Institutes for Biomedical Research (NIBR), Translational Medicine, 4056 Basel, Switzerland
- Institut Pascal, Université Clermont Auvergne, CNRS, Clermont Auvergne INP, 63000 Clermont-Ferrand, France
| | - Philippe Michaud
- Institut Pascal, Université Clermont Auvergne, CNRS, Clermont Auvergne INP, 63000 Clermont-Ferrand, France
| | - Fabrizio Gasparini
- Novartis Institutes for Biomedical Research (NIBR), Translational Medicine, 4056 Basel, Switzerland
| | - Alexandre Avrameas
- Novartis Institutes for Biomedical Research (NIBR), Translational Medicine, 4056 Basel, Switzerland
| |
Collapse
|