1
|
Shao X, Shou Q, Felix K, Ojogho B, Jiang X, Gold BT, Herting MM, Goldwaser EL, Kochunov P, Hong E, Pappas I, Braskie M, Kim H, Cen S, Jann K, Wang DJJ. Age-related decline in blood-brain barrier function is more pronounced in males than females in parietal and temporal regions. eLife 2024; 13:RP96155. [PMID: 39495221 PMCID: PMC11534331 DOI: 10.7554/elife.96155] [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] [Indexed: 11/05/2024] Open
Abstract
The blood-brain barrier (BBB) plays a pivotal role in protecting the central nervous system (CNS), and shielding it from potential harmful entities. A natural decline of BBB function with aging has been reported in both animal and human studies, which may contribute to cognitive decline and neurodegenerative disorders. Limited data also suggest that being female may be associated with protective effects on BBB function. Here, we investigated age and sex-dependent trajectories of perfusion and BBB water exchange rate (kw) across the lifespan in 186 cognitively normal participants spanning the ages of 8-92 years old, using a non-invasive diffusion-prepared pseudo-continuous arterial spin labeling (DP-pCASL) MRI technique. We found that the pattern of BBB kw decline with aging varies across brain regions. Moreover, results from our DP-pCASL technique revealed a remarkable decline in BBB kw beginning in the early 60 s, which was more pronounced in males. In addition, we observed sex differences in parietal and temporal regions. Our findings provide in vivo results demonstrating sex differences in the decline of BBB function with aging, which may serve as a foundation for future investigations into perfusion and BBB function in neurodegenerative and other brain disorders.
Collapse
Affiliation(s)
- Xingfeng Shao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Qinyang Shou
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Kimberly Felix
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Brandon Ojogho
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Xuejuan Jiang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Department of Ophthalmology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Brian T Gold
- Department of Neuroscience, College of Medicine, University of KentuckyFrankfortUnited States
| | - Megan M Herting
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Eric L Goldwaser
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of MedicineBaltimoreUnited States
- Interventional Psychiatry Program, Department of Psychiatry, Weill Cornell MedicineNew YorkUnited States
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Elliot Hong
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Ioannis Pappas
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Meredith Braskie
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Steven Cen
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Kay Jann
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Danny JJ Wang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
2
|
Libri I, Silvestri C, Caratozzolo S, Alberici A, Pilotto A, Archetti S, Trainini L, Borroni B, Padovani A, Benussi A. Association of APOE genotype with blood-brain barrier permeability in neurodegenerative disorders. Neurobiol Aging 2024; 140:33-40. [PMID: 38718740 DOI: 10.1016/j.neurobiolaging.2024.04.003] [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: 01/22/2024] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 06/12/2024]
Abstract
Apolipoprotein E (APOE) is recognized for its role in modulating blood-brain barrier (BBB) permeability in vitro, which may have significant implications for the pathogenesis and progression of neurodegenerative disorders. However, evidence in vivo is contrasting. This study explores the impact of APOE genotypes on BBB integrity among 230 participants experiencing cognitive impairment, encompassing cases of Alzheimer's disease (AD) as well as various non-AD neurodegenerative conditions. To assess BBB integrity, we utilized cerebrospinal fluid (CSF)/serum albumin ratios and CSF/serum kappa and lambda free light chains (FLCs) as indirect markers. Our findings show a dose-dependent increase in BBB permeability in individuals carrying the APOE ε4 allele, marked by elevated CSF/serum albumin and FLCs ratios, with this trend being especially pronounced in AD patients. These results highlight the association of APOE ε4 with BBB permeability, providing valuable insights into the pathophysiology of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ilenia Libri
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Chiara Silvestri
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Salvatore Caratozzolo
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili, Brescia, Italy
| | - Antonella Alberici
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili, Brescia, Italy
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili, Brescia, Italy
| | - Silvana Archetti
- Biotechnology Laboratory and Department of Diagnostics, ASST Spedali Civili, Brescia, Italy
| | - Laura Trainini
- Biotechnology Laboratory and Department of Diagnostics, ASST Spedali Civili, Brescia, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Cognitive and Behavioral Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili, Brescia, Italy; Brain Health Center, University of Brescia, Brescia, Italy
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Neurology Clinic, Department of Medicine, Surgery and Health Sciences, Trieste University Hospital, Trieste, Italy.
| |
Collapse
|
3
|
Campana M, Yakimov V, Moussiopoulou J, Maurus I, Löhrs L, Raabe F, Jäger I, Mortazavi M, Benros ME, Jeppesen R, Meyer Zu Hörste G, Heming M, Giné-Servén E, Labad J, Boix E, Lennox B, Yeeles K, Steiner J, Meyer-Lotz G, Dobrowolny H, Malchow B, Hansen N, Falkai P, Siafis S, Leucht S, Halstead S, Warren N, Siskind D, Strube W, Hasan A, Wagner E. Association of symptom severity and cerebrospinal fluid alterations in recent onset psychosis in schizophrenia-spectrum disorders - An individual patient data meta-analysis. Brain Behav Immun 2024; 119:353-362. [PMID: 38608742 DOI: 10.1016/j.bbi.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
Neuroinflammation and blood-cerebrospinal fluid barrier (BCB) disruption could be key elements in schizophrenia-spectrum disorderś(SSDs) etiology and symptom modulation. We present the largest two-stage individual patient data (IPD) meta-analysis, investigating the association of BCB disruption and cerebrospinal fluid (CSF) alterations with symptom severity in first-episode psychosis (FEP) and recent onset psychotic disorder (ROP) individuals, with a focus on sex-related differences. Data was collected from PubMed and EMBASE databases. FEP, ROP and high-risk syndromes for psychosis IPD were included if routine basic CSF-diagnostics were reported. Risk of bias of the included studies was evaluated. Random-effects meta-analyses and mixed-effects linear regression models were employed to assess the impact of BCB alterations on symptom severity. Published (6 studies) and unpublished IPD from n = 531 individuals was included in the analyses. CSF was altered in 38.8 % of individuals. No significant differences in symptom severity were found between individuals with and without CSF alterations (SMD = -0.17, 95 %CI -0.55-0.22, p = 0.341). However, males with elevated CSF/serum albumin ratios or any CSF alteration had significantly higher positive symptom scores than those without alterations (SMD = 0.34, 95 %CI 0.05-0.64, p = 0.037 and SMD = 0.29, 95 %CI 0.17-0.41p = 0.005, respectively). Mixed-effects and simple regression models showed no association (p > 0.1) between CSF parameters and symptomatic outcomes. No interaction between sex and CSF parameters was found (p > 0.1). BCB disruption appears highly prevalent in early psychosis and could be involved in positive symptomś severity in males, indicating potential difficult-to-treat states. This work highlights the need for considering BCB breakdownand sex-related differences in SSDs clinical trials and treatment strategies.
Collapse
Affiliation(s)
- Mattia Campana
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany.
| | - Vladislav Yakimov
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Joanna Moussiopoulou
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Isabel Maurus
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Lisa Löhrs
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Florian Raabe
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany
| | - Iris Jäger
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Matin Mortazavi
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany
| | - Michael E Benros
- Copenhagen Research Centre for Biological and Precision Psychiatry. Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rose Jeppesen
- Copenhagen Research Centre for Biological and Precision Psychiatry. Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Michael Heming
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Eloi Giné-Servén
- Department of Psychiatry, Hospital Universitario de Navarra, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Javier Labad
- Department of Mental Health, Hospital de Mataró, Consorci Sanitari del Maresme, Mataró, Spain; Translational Neuroscience Research Unit I3PT-INc-UAB, Institut de Innovació i Investigació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - Ester Boix
- Department of Mental Health, Hospital de Mataró, Consorci Sanitari del Maresme, Mataró, Spain
| | - Belinda Lennox
- Department of Psychiatry, University of Oxford and Oxford Health NHS Foundation Trust, Oxford, UK
| | - Ksenija Yeeles
- Department of Psychiatry, University of Oxford and Oxford Health NHS Foundation Trust, Oxford, UK
| | - Johann Steiner
- Department of Psychiatry, Magdeburg University Hospital, Magdeburg, Germany
| | | | - Henrik Dobrowolny
- Department of Psychiatry, Magdeburg University Hospital, Magdeburg, Germany
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany; DZPG (German Center for Mental Health), partner site München/Augsburg, Germany
| | - Spyridon Siafis
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University Munich, Munich, Germany
| | - Stefan Leucht
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University Munich, Munich, Germany
| | - Sean Halstead
- Department of Psychiatry, School of Medicine, University of Queensland, Brisbane, Australia
| | - Nicola Warren
- Department of Psychiatry, School of Medicine, University of Queensland, Brisbane, Australia
| | - Dan Siskind
- Department of Psychiatry, School of Medicine, University of Queensland, Brisbane, Australia
| | - Wolfgang Strube
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany; DZPG (German Center for Mental Health), partner site München/Augsburg, Germany
| | - Elias Wagner
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany; Evidence-based Psychiatry and Psychotherapy, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| |
Collapse
|
4
|
Shao X, Shou Q, Felix K, Ojogho B, Jiang X, Gold BT, Herting MM, Goldwaser EL, Kochunov P, Hong LE, Pappas I, Braskie M, Kim H, Cen S, Jann K, Wang DJJ. Age-Related Decline in Blood-Brain Barrier Function is More Pronounced in Males than Females in Parietal and Temporal Regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575463. [PMID: 38293052 PMCID: PMC10827081 DOI: 10.1101/2024.01.12.575463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The blood-brain barrier (BBB) plays a pivotal role in protecting the central nervous system (CNS), shielding it from potential harmful entities. A natural decline of BBB function with aging has been reported in both animal and human studies, which may contribute to cognitive decline and neurodegenerative disorders. Limited data also suggest that being female may be associated with protective effects on BBB function. Here we investigated age and sex-dependent trajectories of perfusion and BBB water exchange rate (kw) across the lifespan in 186 cognitively normal participants spanning the ages of 8 to 92 years old, using a non-invasive diffusion prepared pseudo-continuous arterial spin labeling (DP-pCASL) MRI technique. We found that the pattern of BBB kw decline with aging varies across brain regions. Moreover, results from our DP-pCASL technique revealed a remarkable decline in BBB kw beginning in the early 60s, which was more pronounced in males. In addition, we observed sex differences in parietal and temporal regions. Our findings provide in vivo results demonstrating sex differences in the decline of BBB function with aging, which may serve as a foundation for future investigations into perfusion and BBB function in neurodegenerative and other brain disorders.
Collapse
Affiliation(s)
- Xingfeng Shao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Qinyang Shou
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Kimberly Felix
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - Brandon Ojogho
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Xuejuan Jiang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
- Department of Ophthalmology, Keck School of Medicine, University of Southern California
| | - Brian T. Gold
- Department of Neuroscience, College of Medicine, University of Kentucky
| | - Megan M Herting
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - Eric L Goldwaser
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine
- Interventional Psychiatry Program, Department of Psychiatry, Weill Cornell Medicine
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston
| | - L. Elliot Hong
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Ioannis Pappas
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Meredith Braskie
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Steven Cen
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern California
| | - Kay Jann
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Danny JJ Wang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern California
| |
Collapse
|
5
|
Candeloro R, Ferri C, Bellini T, Pugliatti M, Castellazzi M. Breaking Barriers: Unveiling Sex-Related Differences in Cerebrospinal Fluid Analysis-A Narrative Review. BIOLOGY 2024; 13:420. [PMID: 38927300 PMCID: PMC11200519 DOI: 10.3390/biology13060420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
(1) Background: The recent emphasis on sexual and gender diversity's impact on human health underscores the need for tailored diagnostic and therapeutic approaches in neurology. The aim of this article is to conduct a narrative review of the available scientific literature on sex differences in cerebrospinal fluid analysis. (2) Methods: The literature search encompassed PubMed databases, focusing on cerebrospinal fluid analysis and sex differences, considering parameters like cerebrospinal fluid protein content, cell count, albumin quotient (QAlb) and intrathecal IgG synthesis. (3) Results: Nine articles from the past two decades were identified, revealing limited research in this area. Males consistently exhibited higher cerebrospinal fluid protein content and albumin quotient values across various pathologies and age groups. Consequently, males more frequently manifested blood-cerebrospinal fluid barrier dysfunction than females. No significant sex differences were observed in cerebrospinal fluid leukocyte count or intrathecal IgG synthesis. (4) Conclusions: This review highlights the dearth of research on sex differences in cerebrospinal fluid analysis, despite consistent findings of higher protein content and albumin quotient values in males. Revisiting current diagnostic thresholds based on sex is crucial for accurate prognosis and personalised treatment strategies in neurological disorders. Moving towards sex-specific approaches in clinical practice is imperative for advancing personalised medicine.
Collapse
Affiliation(s)
- Raffaella Candeloro
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (T.B.); (M.P.)
| | - Caterina Ferri
- Department of Neuroscience, “S. Anna” University Hospital, 44124 Ferrara, Italy;
| | - Tiziana Bellini
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (T.B.); (M.P.)
- University Strategic Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Maura Pugliatti
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (T.B.); (M.P.)
| | - Massimiliano Castellazzi
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (T.B.); (M.P.)
- University Strategic Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
6
|
Noguchi Y, Asano H, Masuda R, Teshigawara Y, Go M, Kimura M, Usami E, Yoshimura T. Relationship between Anaplastic Lymphoma Kinase Inhibitors and Epileptic Seizure Disorder: A Post-Marketing Surveillance Study. Oncology 2024; 102:996-1003. [PMID: 38768582 DOI: 10.1159/000539426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/16/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Anaplastic lymphoma kinase (ALK) has been to be involved in the uptake and regulation of dopamine 2 receptor (D2R), a G protein-coupled receptor expressed in various brain regions. Therefore, it is crucial to understand the relationship between ALK inhibitors and seizures is an important issue. This study investigated the relationship between ALK inhibitors and seizures. METHODS This study investigated the relationship between ALK inhibitors and seizures through a disproportionality analysis using the US Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS). The target drugs were the ALK inhibitors crizotinib, ceritinib, alectinib, brigatinib, and lorlatinib. The seizures covered were defined high-level group term (HLGT): "Seizures (incl. subtype)" including high-level term (HLT): "seizures and seizure disorders NEC." This study used the information component (IC), a signal score, as a Bayesian statistical method for disproportionality analysis. The signal detection criteria used in this study were the same as those reported previously: a lower limit of 95% credible interval (CrI) for IC >0. RESULTS The signal scores of '"seizures and seizure disorders not elsewhere classified (NEC)" "for each ALK inhibitor were crizotinib (IC: -0.00052, 95% CrI: -0.38-0.27), ceritinib (IC: 1.18, 95% CrI: 0.68-1.54), alectinib (IC: 0.68, 95% CrI: 0.19-1.02), brigatinib (IC: 1.04, 95% CrI: 0.32-1.54), and lorlatinib (IC: 0.82, 95% CrI: 0.11-1.32). On the other hand, "generalized tonic-clonic seizures," "partial simple seizures NEC," "absence seizures," and "partial complex seizures" had no or few reported cases, and no signal was detected. CONCLUSION To our knowledge, this is the first report to evaluate the relationship between ALK inhibitors and seizures using post-marketing surveillance data. These results suggest that ceritinib, alectinib, brigatinib, and lorlatinib, which are highly brain-migrating drugs, are associated with seizures.
Collapse
Affiliation(s)
- Yoshihiro Noguchi
- Laboratory of Clinical Pharmacy, Gifu Pharmaceutical University, Gifu, Japan
- Laboratory of Medical Collaborative Pharmacy, Gifu Pharmaceutical University, Gifu, Japan
| | - Hiroki Asano
- Department of Pharmacy, Ogaki Municipal Hospital, Gifu, Japan
| | - Rikuto Masuda
- Laboratory of Clinical Pharmacy, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuta Teshigawara
- Laboratory of Clinical Pharmacy, Gifu Pharmaceutical University, Gifu, Japan
| | - Makiko Go
- Department of Pharmacy, Ogaki Municipal Hospital, Gifu, Japan
| | - Michio Kimura
- Laboratory of Clinical Pharmacy, Gifu Pharmaceutical University, Gifu, Japan
- Department of Pharmacy, Ogaki Municipal Hospital, Gifu, Japan
| | - Eiseki Usami
- Laboratory of Medical Collaborative Pharmacy, Gifu Pharmaceutical University, Gifu, Japan
- Department of Pharmacy, Ogaki Municipal Hospital, Gifu, Japan
| | - Tomoaki Yoshimura
- Laboratory of Clinical Pharmacy, Gifu Pharmaceutical University, Gifu, Japan
- Laboratory of Medical Collaborative Pharmacy, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
7
|
Wang YT, Therriault J, Servaes S, Tissot C, Rahmouni N, Macedo AC, Fernandez-Arias J, Mathotaarachchi SS, Benedet AL, Stevenson J, Ashton NJ, Lussier FZ, Pascoal TA, Zetterberg H, Rajah MN, Blennow K, Gauthier S, Rosa-Neto P. Sex-specific modulation of amyloid-β on tau phosphorylation underlies faster tangle accumulation in females. Brain 2024; 147:1497-1510. [PMID: 37988283 PMCID: PMC10994548 DOI: 10.1093/brain/awad397] [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: 08/10/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/23/2023] Open
Abstract
Females are disproportionately affected by dementia due to Alzheimer's disease. Despite a similar amyloid-β (Aβ) load, a higher load of neurofibrillary tangles (NFTs) is seen in females than males. Previous literature has proposed that Aβ and phosphorylated-tau (p-tau) synergism accelerates tau tangle formation, yet the effect of biological sex in this process has been overlooked. In this observational study, we examined longitudinal neuroimaging data from the TRIAD and ADNI cohorts from Canada and USA, respectively. We assessed 457 participants across the clinical spectrum of Alzheimer's disease. All participants underwent baseline multimodal imaging assessment, including MRI and PET, with radioligands targeting Aβ plaques and tau tangles, respectively. CSF data were also collected. Follow-up imaging assessments were conducted at 1- and 2-year intervals for the TRIAD cohort and 1-, 2- and 4-year intervals for the ADNI cohort. The upstream pathological events contributing to faster tau progression in females were investigated-specifically, whether the contribution of Aβ and p-tau synergism to accelerated tau tangle formation is modulated by biological sex. We hypothesized that cortical Aβ predisposes tau phosphorylation and tangle accumulation in a sex-specific manner. Findings revealed that Aβ-positive females presented higher CSF p-tau181 concentrations compared with Aβ-positive males in both the TRIAD (P = 0.04, Cohen's d = 0.51) and ADNI (P = 0.027, Cohen's d = 0.41) cohorts. In addition, Aβ-positive females presented faster NFT accumulation compared with their male counterparts (TRIAD: P = 0.026, Cohen's d = 0.52; ADNI: P = 0.049, Cohen's d = 1.14). Finally, the triple interaction between female sex, Aβ and CSF p-tau181 was revealed as a significant predictor of accelerated tau accumulation at the 2-year follow-up visit (Braak I: P = 0.0067, t = 2.81; Braak III: P = 0.017, t = 2.45; Braak IV: P = 0.002, t = 3.17; Braak V: P = 0.006, t = 2.88; Braak VI: P = 0.0049, t = 2.93). Overall, we report sex-specific modulation of cortical Aβ in tau phosphorylation, consequently facilitating faster NFT progression in female individuals over time. This presents important clinical implications and suggests that early intervention that targets Aβ plaques and tau phosphorylation may be a promising therapeutic strategy in females to prevent the further accumulation and spread of tau aggregates.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Sulantha S Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, 4011 Stavanger, Norway
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London SE5 9RX, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London SE5 8AF, UK
| | - Firoza Z Lussier
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, 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
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
- Montreal Neurological Institute, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
8
|
Castellazzi M, Candeloro R, Trevisan C, Permunian S, Buscemi G, Ghisellini S, Negri G, Gilli G, Ferri C, Bellini T, Pizzicotti S, Pugliatti M. Sex Differences in Albumin Quotient and Cerebrospinal Fluid Total Protein Content Do Not Depend on Anthropometric Factors. J Pers Med 2024; 14:362. [PMID: 38672989 PMCID: PMC11051272 DOI: 10.3390/jpm14040362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
(1) Background: Cerebrospinal fluid (CSF)/serum albumin quotient (QAlb) and CSF total protein (TP) are more elevated in males than females, and this has been hypothesised to be due to anthropometric differences between the sexes. This study aimed to investigate QAlb and CSF TP as a function of body height, weight, and body mass index (BMI). (2) Methods: A total of 207 patients were included in the study and analysed blinded to clinical diagnosis. (3) Results: Multivariable linear regressions were run to predict log-transformed Qalb and log-transformed CSF TP value from age, sex, weight, and height (first model) or from age, sex, and BMI (second model). In both models, age (β = 0.004, 95% CI = 0.002 to 0.006) and sex (β = -0.095, 95% CI = -0.169 to -0.021, and β = -0.135, 95% CI = -0.191 to -0.079) were significant predictors for QAlb, but weight, height, and BMI were not. Similarly, age (β = 0.004, 95% CI = 0.003 to 0.006) and sex (β = -0.077, 95% CI = -0.142 to -0.013, and β = -0.109, 95% CI = -0.157 to -0.060) were significant predictors for CSF TP, while anthropometric characteristics were not. No differences in QAlb and CSF TP were found when grouping males and females by BMI status. (4) Conclusions: Our data suggest that anthropometric characteristics could not explain the sex-related differences in QAlb and CSF TP.
Collapse
Affiliation(s)
- Massimiliano Castellazzi
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (S.P.); (G.B.); (G.G.); (T.B.); (M.P.)
- University Strategic Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Raffaella Candeloro
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (S.P.); (G.B.); (G.G.); (T.B.); (M.P.)
| | - Caterina Trevisan
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Samantha Permunian
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (S.P.); (G.B.); (G.G.); (T.B.); (M.P.)
| | - Gaia Buscemi
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (S.P.); (G.B.); (G.G.); (T.B.); (M.P.)
| | - Sara Ghisellini
- Chemical-Clinical Analysis Laboratory, “S. Anna” University Hospital, 44124 Ferrara, Italy; (S.G.); (G.N.); (S.P.)
| | - Giovanna Negri
- Chemical-Clinical Analysis Laboratory, “S. Anna” University Hospital, 44124 Ferrara, Italy; (S.G.); (G.N.); (S.P.)
| | - Giada Gilli
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (S.P.); (G.B.); (G.G.); (T.B.); (M.P.)
| | - Caterina Ferri
- Department of Neuroscience, “S. Anna” University Hospital, 44124 Ferrara, Italy;
| | - Tiziana Bellini
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (S.P.); (G.B.); (G.G.); (T.B.); (M.P.)
- University Strategic Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Stefano Pizzicotti
- Chemical-Clinical Analysis Laboratory, “S. Anna” University Hospital, 44124 Ferrara, Italy; (S.G.); (G.N.); (S.P.)
| | - Maura Pugliatti
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (R.C.); (S.P.); (G.B.); (G.G.); (T.B.); (M.P.)
| |
Collapse
|
9
|
Rademeyer KM, R Nass S, Jones AM, Ohene-Nyako M, Hauser KF, McRae M. Fentanyl dysregulates neuroinflammation and disrupts blood-brain barrier integrity in HIV-1 Tat transgenic mice. J Neurovirol 2024; 30:1-21. [PMID: 38280928 PMCID: PMC11232468 DOI: 10.1007/s13365-023-01186-4] [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: 06/17/2023] [Revised: 10/29/2023] [Accepted: 11/16/2023] [Indexed: 01/29/2024]
Abstract
Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and dysregulate an already compromised inflammatory response. Despite the rise in fentanyl abuse and the close association between opioid abuse and HIV infection, the interactive comorbidity between fentanyl abuse and HIV has yet to be examined in vivo. The HIV-1 Tat-transgenic mouse model was used to understand the interactive effects between fentanyl and HIV. Tat is an essential protein produced during HIV that drives the transcription of new virions and exerts neurotoxic effects within the brain. The Tat-transgenic mouse model uses a glial fibrillary acidic protein (GFAP)-driven tetracycline promoter which limits Tat production to the brain and this model is well used for examining mechanisms related to neuroHIV. After 7 days of fentanyl exposure, brains were harvested. Tight junction proteins, the vascular cell adhesion molecule, and platelet-derived growth factor receptor-β were measured to examine the integrity of the blood brain barrier. The immune response was assessed using a mouse-specific multiplex chemokine assay. For the first time in vivo, we demonstrate that fentanyl by itself can severely disrupt the blood-brain barrier and dysregulate the immune response. In addition, we reveal associations between inflammatory markers and tight junction proteins at the blood-brain barrier.
Collapse
Affiliation(s)
- Kara M Rademeyer
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Austin M Jones
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - MaryPeace McRae
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, 22908, U.S.A..
| |
Collapse
|
10
|
Seeliger T, Gingele S, Güzeloglu YE, Heitmann L, Lüling B, Kohle F, Preßler H, Stascheit F, Motte J, Fisse AL, Grüter T, Pitarokoili K, Skripuletz T. Comparative analysis of albumin quotient and total CSF protein in immune-mediated neuropathies: a multicenter study on diagnostic implications. Front Neurol 2024; 14:1330484. [PMID: 38264088 PMCID: PMC10803547 DOI: 10.3389/fneur.2023.1330484] [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: 10/31/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction Blood-cerebrospinal fluid (CSF) barrier dysfunction is pivotal for diagnosing immune-mediated neuropathies, especially in spinal nerve root inflammation. Typically, either total CSF protein or the CSF to serum albumin ratio (QAlb) is measured. Total CSF protein measurements have limitations, notably its fixed reference value regardless of age, in contrast to the age-dependent reference for QAlb. Our goal was to evaluate both markers in patients with immune-mediated neuropathies. Methods In our multicenter research, we collected retrospective CSF data from patients suffering from immune-mediated neuropathies across four German research centers. These parameters were analyzed in relation to their clinical characteristics. Results Out of 419 samples, 36 (8.6%) displayed a notable variation between total CSF protein and QAlb values. A detailed analysis revealed that patients displaying elevated QAlb but normal total CSF protein levels were significantly younger at disease onset (p = 0.01), at the time of diagnosis (p = 0.005), and when undergoing lumbar puncture (p = 0.001) compared to patients with elevated CSF protein and normal QAlb levels. These effects were especially evident for the subgroup of samples derived by female patients. Discussion Our work confirms the crucial role of QAlb in diagnosing immune-mediated neuropathies and particularly its efficacy as a marker for evaluating the blood-CSF barrier in patients with an earlier disease onset. Considering the significance of the albumin quotient, its assessment is especially advisable in younger patients of female sex to avoid missing a potential barrier dysfunction that might be falsely negative when using total protein.
Collapse
Affiliation(s)
- Tabea Seeliger
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Stefan Gingele
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | - Lena Heitmann
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Benjamin Lüling
- Department of Neurology, St Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Felix Kohle
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Hannah Preßler
- Department of Neurology with Experimental Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität, Berlin, Germany
- Neuroscience Clinical Research Center, Charité — Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frauke Stascheit
- Department of Neurology with Experimental Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität, Berlin, Germany
- Neuroscience Clinical Research Center, Charité — Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jeremias Motte
- Department of Neurology, St Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Anna Lena Fisse
- Department of Neurology, St Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Thomas Grüter
- Department of Neurology, St Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Kalliopi Pitarokoili
- Department of Neurology, St Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | | |
Collapse
|
11
|
Puig‐Pijoan A, Jimenez‐Balado J, Fernández‐Lebrero A, García‐Escobar G, Navalpotro‐Gómez I, Contador J, Manero‐Borràs R, Puente‐Periz V, Suárez A, Muñoz FJ, Grau‐Rivera O, Suárez‐Calvet M, de la Torre R, Roquer J, Ois A. Risk of cognitive decline progression is associated to increased blood-brain-barrier permeability: A longitudinal study in a memory unit clinical cohort. Alzheimers Dement 2024; 20:538-548. [PMID: 37727082 PMCID: PMC10916969 DOI: 10.1002/alz.13433] [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: 05/11/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/21/2023]
Abstract
INTRODUCTION This study examined the relationship between blood-brain-barrier permeability (BBBp), measured by cerebrospinal fluid/serum albumin ratio (QAlb), and cognitive decline progression in a clinical cohort. METHODS This prospective observational study included 334 participants from the BIODEGMAR cohort. Cognitive decline progression was defined as an increase in Global Deterioration Scale and/or Clinical Dementia Rating scores. Associations between BBBp, demographics, and clinical factors were explored. RESULTS Male sex, diabetes mellitus, and cerebrovascular burden were associated with increased log-QAlb. Vascular cognitive impairment patients had the highest log-QAlb levels. Among the 273 participants with valid follow-up data, 154 (56.4%) showed cognitive decline progression. An 8% increase in the hazard of clinical worsening was observed for each 10% increase in log-QAlb. DISCUSSION These results suggest that increased BBBp in individuals with cognitive decline may contribute to clinical worsening, pointing to potential targeted therapies. QAlb could be a useful biomarker for identifying patients with a worse prognosis.
Collapse
|
12
|
Zheng HT, Wu Z, Mielke MM, Murray AM, Ryan J. Plasma Biomarkers of Alzheimer's Disease and Neurodegeneration According to Sociodemographic Characteristics and Chronic Health Conditions. J Prev Alzheimers Dis 2024; 11:1189-1197. [PMID: 39350363 PMCID: PMC11436401 DOI: 10.14283/jpad.2024.142] [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: 05/21/2024] [Accepted: 06/23/2024] [Indexed: 10/04/2024]
Abstract
Ultrasensitive assays have been developed which enable biomarkers of Alzheimer's disease pathology and neurodegeneration to be measured in blood. These biomarkers can aid in diagnosis, and have been used to predict risk of cognitive decline and Alzheimer's disease. The ease and cost-effectiveness of blood collections means that these biomarkers could be applied more broadly in population-based screening, however it is critical to first understand what other factors could affect blood biomarker levels. The aim of this review was to determine the extent that sociodemographic, lifestyle and health factors have been associated with blood biomarkers of Alzheimer's disease and neuropathology. Of the 32 studies included in this review, all but one measured biomarker levels in plasma, and age and sex were the most commonly investigated factors. The most consistent significant findings were a positive association between age and neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP), and females had higher GFAP than men. Apolipoprotein ε4 allele carriers had lower Aβ42 and Aβ42/40 ratio. Body mass index was negatively associated with GFAP and NfL, and chronic kidney disease with higher levels of all biomarkers. Too few studies have investigated other chronic health conditions and this requires further investigation. Given the potential for plasma biomarkers to enhance Alzheimer's disease diagnosis in primary care, it is important to understand how to interpret the biomarkers in light of factors that physiologically impact blood biomarker levels. This information will be critical for the establishment of reference ranges and thus the correct interpretation of these biomarkers in clinical screening.
Collapse
Affiliation(s)
- H T Zheng
- Joanne Ryan, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia,
| | | | | | | | | |
Collapse
|
13
|
Cheng X, Li Y, Wang Y, Sun Y, Lian Y. Impact of blood-brain barrier disruption on clinical features and treatment response in patients with newly diagnosed autoimmune encephalitis. J Neuroimmunol 2023; 383:578203. [PMID: 37740995 DOI: 10.1016/j.jneuroim.2023.578203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
AIMS Blood-brain barrier (BBB) breakdown is an essential mechanism in inflammatory diseases of the central nervous system. However, the association between BBB integrity and autoimmune encephalitis (AE) has not been investigated. Our study aimed to analyze this relationship in patients with AE between BBB integrity with clinical manifestations and therapeutic responses. METHODS Our study enrolled 147 patients with AE who were newly diagnosed at the First Affiliated Hospital of Zhengzhou University between August 2015 and December 2021. Patients were classified into normal or damaged BBB groups based on cerebrospinal fluid (CSF) albumin/serum albumin (QAlb). To evaluate the severity of the illness, we used the modified Rankin Scale (mRS) and the Clinical Assessment Scale for Autoimmune Encephalitis (CASE). RESULTS We found a higher proportion of males, higher CSF protein, immunoglobulin IgG, and 24-h intrathecal IgG synthesis rate in the damaged BBB group. The improvement rate was lower in the damaged BBB group, but we found that double- or triple-combination immunotherapy had better clinical outcomes than single immunotherapy. In addition, there was a positive correlation between the CASE score and mRS score, and a positive correlation between the CASE score or mRS score and QAlb on admission. CONCLUSIONS BBB integrity is closely related to the clinical features and treatment responses of newly diagnosed AE. Patients with AE and a damaged BBB may benefit from combination immunotherapy.
Collapse
Affiliation(s)
- Xuan Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yimeng Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaoyao Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yidi Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Bellaver B, Puig-Pijoan A, Ferrari-Souza JP, Leffa DT, Lussier FZ, Ferreira PCL, Tissot C, Povala G, Therriault J, Benedet AL, Ashton NJ, Servaes S, Chamoun M, Stevenson J, Rahmouni N, Vermeiren M, Macedo AC, Fernández-Lebrero A, García-Escobar G, Navalpotro-Gómez I, Lopez O, Tudorascu DL, Cohen A, Villemagne VL, Klunk WE, Gauthier S, Zimmer ER, Karikari TK, Blennow K, Zetterberg H, Suárez-Calvet M, Rosa-Neto P, Pascoal TA. Blood-brain barrier integrity impacts the use of plasma amyloid-β as a proxy of brain amyloid-β pathology. Alzheimers Dement 2023; 19:3815-3825. [PMID: 36919582 PMCID: PMC10502181 DOI: 10.1002/alz.13014] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/08/2022] [Accepted: 01/25/2023] [Indexed: 03/16/2023]
Abstract
INTRODUCTION Amyloid-β (Aβ) and tau can be quantified in blood. However, biological factors can influence the levels of brain-derived proteins in the blood. The blood-brain barrier (BBB) regulates protein transport between cerebrospinal fluid (CSF) and blood. BBB altered permeability might affect the relationship between brain and blood biomarkers. METHODS We assessed 224 participants in research (TRIAD, n = 96) and clinical (BIODEGMAR, n = 128) cohorts with plasma and CSF/positron emission tomography Aβ, p-tau, and albumin measures. RESULTS Plasma Aβ42/40 better identified CSF Aβ42/40 and Aβ-PET positivity in individuals with high BBB permeability. An interaction between plasma Aβ42/40 and BBB permeability on CSF Aβ42/40 was observed. Voxel-wise models estimated that the association of positron emission tomography (PET), with plasma Aβ was most affected by BBB permeability in AD-related brain regions. BBB permeability did not significantly impact the relationship between brain and plasma p-tau levels. DISCUSSION These findings suggest that BBB integrity may influence the performance of plasma Aβ, but not p-tau, biomarkers in research and clinical settings. HIGHLIGHTS BBB permeability affects the association between brain and plasma Aβ levels. BBB integrity does not affect the association between brain and plasma p-tau levels. Plasma Aβ was most affected by BBB permeability in AD-related brain regions. BBB permeability increases with age but not according to cognitive status.
Collapse
Affiliation(s)
- Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Albert Puig-Pijoan
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Douglas T Leffa
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Firoza Z Lussier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Pamela C L Ferreira
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Guilherme Povala
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Aida Fernández-Lebrero
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | | | - Irene Navalpotro-Gómez
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Oscar Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dana L Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ann Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Marc Suárez-Calvet
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
Jullienne A, Szu JI, Quan R, Trinh MV, Norouzi T, Noarbe BP, Bedwell AA, Eldridge K, Persohn SC, Territo PR, Obenaus A. Cortical cerebrovascular and metabolic perturbations in the 5xFAD mouse model of Alzheimer's disease. Front Aging Neurosci 2023; 15:1220036. [PMID: 37533765 PMCID: PMC10392850 DOI: 10.3389/fnagi.2023.1220036] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/03/2023] [Indexed: 08/04/2023] Open
Abstract
Introduction The 5xFAD mouse is a popular model of familial Alzheimer's disease (AD) that is characterized by early beta-amyloid (Aβ) deposition and cognitive decrements. Despite numerous studies, the 5xFAD mouse has not been comprehensively phenotyped for vascular and metabolic perturbations over its lifespan. Methods Male and female 5xFAD and wild type (WT) littermates underwent in vivo 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging at 4, 6, and 12 months of age to assess regional glucose metabolism. A separate cohort of mice (4, 8, 12 months) underwent "vessel painting" which labels all cerebral vessels and were analyzed for vascular characteristics such as vessel density, junction density, vessel length, network complexity, number of collaterals, and vessel diameter. Results With increasing age, vessels on the cortical surface in both 5xFAD and WT mice showed increased vessel length, vessel and junction densities. The number of collateral vessels between the middle cerebral artery (MCA) and the anterior and posterior cerebral arteries decreased with age but collateral diameters were significantly increased only in 5xFAD mice. MCA total vessel length and junction density were decreased in 5xFAD mice compared to WT at 4 months. Analysis of 18F-FDG cortical uptake revealed significant differences between WT and 5xFAD mice spanning 4-12 months. Broadly, 5xFAD males had significantly increased 18F-FDG uptake at 12 months compared to WT mice. In most cortical regions, female 5xFAD mice had reduced 18F-FDG uptake compared to WT across their lifespan. Discussion While the 5xFAD mouse exhibits AD-like cognitive deficits as early as 4 months of age that are associated with increasing Aβ deposition, we only found significant differences in cortical vascular features in males, not in females. Interestingly, 5xFAD male and female mice exhibited opposite effects in 18F-FDG uptake. The MCA supplies blood to large portions of the somatosensory cortex and portions of motor and visual cortex and increased vessel length alongside decreased collaterals which coincided with higher metabolic rates in 5xFAD mice. Thus, a potential mismatch between metabolic demand and vascular delivery of nutrients in the face of increasing Aβ deposition could contribute to the progressive cognitive deficits seen in the 5xFAD mouse model.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Jenny I. Szu
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Ryan Quan
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Michelle V. Trinh
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Tannoz Norouzi
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Brenda P. Noarbe
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Amanda A. Bedwell
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Kierra Eldridge
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Scott C. Persohn
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Paul R. Territo
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
16
|
Alarcan H, Vourc'h P, Berton L, Benz-De Bretagne I, Piver E, Andres CR, Corcia P, Veyrat-Durebex C, Blasco H. Implication of Central Nervous System Barrier Impairment in Amyotrophic Lateral Sclerosis: Gender-Related Difference in Patients. Int J Mol Sci 2023; 24:11196. [PMID: 37446372 DOI: 10.3390/ijms241311196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Central nervous system (CNS) barrier impairment has been reported in amyotrophic lateral sclerosis (ALS), highlighting its potential significance in the disease. In this context, we aim to shed light on its involvement in the disease, by determining albumin quotient (QAlb) at the time of diagnosis of ALS in a large cohort of patients. Patients from the university hospital of Tours (n = 307) were included in this monocentric, retrospective study. In total, 92 patients (30%) had elevated QAlb levels. This percentage was higher in males (43%) than in females (15%). Interestingly, QAlb was not associated with age of onset, age at sampling or diagnostic delay. However, we found an association with ALS functional rating scale-revised (ALSFRS-r) at diagnosis but this was significant only in males. The QAlb levels were not linked to the presence of a pathogenic mutation. Finally, we performed a multivariate survival analysis and found that QAlb was significantly associated with survival in male patients (HR = 2.3, 95% CI = 1.2-4.3, p = 0.009). A longitudinal evaluation of markers of barrier impairment, in combination with inflammatory biomarkers, could give insight into the involvement of CNS barrier impairment in the pathogenesis of the disease. The gender difference might guide the development of new drugs and help personalise the treatment of ALS.
Collapse
Affiliation(s)
- Hugo Alarcan
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Patrick Vourc'h
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Lise Berton
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
| | - Isabelle Benz-De Bretagne
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
| | - Eric Piver
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
| | - Christian R Andres
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Philippe Corcia
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
- Service de Neurologie, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
| | - Charlotte Veyrat-Durebex
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Hélène Blasco
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| |
Collapse
|
17
|
Schellin J, Neumann A, Schramm P, Laabs BH, Küchler J, Schacht H. Reversible intracranial contrast medium accumulation after embolization of unruptured cerebral aneurysms and its association with transient neurological deficits: A single center experience. Brain Circ 2023; 9:178-184. [PMID: 38020948 PMCID: PMC10679634 DOI: 10.4103/bc.bc_25_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/10/2023] [Accepted: 06/18/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Use of iodine-containing contrast medium (CM) is obligatory for endovascular treatment (EVT) of cerebral aneurysms. After EVT, intracranial density increases (DIs) can be detected in cranial computed tomography (CT). Those DI can correspond to subarachnoid hemorrhage (SAH), infarction or reversible CM accumulation (RCMA). The latter can be mistaken for hemorrhage, especially if they are accompanied by neurological deficits. OBJECTIVE To analyze postinterventional DI after EVT of unruptured cerebral aneurysms and associated clinical symptoms and to identify risk factors for the occurrence of RCMA. METHODS For differentiation of DI, we compared CT scans following EVT and additionally 24 h ± 5 h later. Diagnosis of RCMA was based on marked regression of DI on follow-up scans. We analyzed continuous variables (age, duration of intervention and anesthesia, aneurysm diameter, amount of CM and renal function) and categorial variables (gender, aneurysm location, devices for EVT, antiplatelet therapy [APT] and associated neurological deficits) to identify risk factors for the occurrence of RCMA. RESULTS We studied 58 patients (44 female, mean age 59.5 [range 39-81]) who underwent EVT for a total of 68 cerebral aneurysms in 62 therapy sessions over a 3-year period without periprocedural complications. Postinterventional DI occurred after 17 therapy sessions. All 17 DI turned out to be RCMA in the follow-up imaging. Two patients who had no DI on initial postinterventional CT showed new SAH on follow-up CT. Infarctions were not observed. Transient neurological deficits occurred in eight patients (12.9%) and were associated with RCMA (P = 0.010). Postinterventional RCMA was associated with the duration of EVT (P = 0.038) and with APT (acetylsalicylic acid [ASA] + clopidogrel: P =0.040; ASA alone: P =0.011). CONCLUSIONS RCMA is common after EVT of unruptured cerebral aneurysms and often accompanied by transient neurological deficits. Long procedure duration and APT appear to predispose to the occurrence of RCMA.
Collapse
Affiliation(s)
- Jenna Schellin
- Department of Neuroradiology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Alexander Neumann
- Department of Neuroradiology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Peter Schramm
- Department of Neuroradiology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Björn-Hergen Laabs
- Institute of Medical Biometry and Statistics (I. R. K.), University of Lübeck, Lübeck, Germany
| | - Jan Küchler
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Hannes Schacht
- Department of Neuroradiology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
18
|
Musaeus CS, Gleerup HS, Hasselbalch SG, Waldemar G, Simonsen AH. Progression of Blood-Brain Barrier Leakage in Patients with Alzheimer's Disease as Measured with the Cerebrospinal Fluid/Plasma Albumin Ratio Over Time. J Alzheimers Dis Rep 2023; 7:535-541. [PMID: 37313491 PMCID: PMC10259070 DOI: 10.3233/adr-230016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/23/2023] [Indexed: 06/15/2023] Open
Abstract
Background Studies have found a disruption of the blood-brain barrier (BBB) in patients with Alzheimer's disease (AD), but there is little evidence of the changes in the BBB over time. The cerebrospinal fluid's (CSF) protein concentration can be used as an indirect measurement for the permeability of the BBB using the CSF/plasma albumin quotient (Q-Alb) or total CSF protein. Objective In the current study, we wanted to investigate the changes in Q-Alb in patients with AD over time. Methods A total of 16 patients diagnosed with AD, who had at least two lumbar punctures performed, were included in the current study. Results The difference in Q-Alb over time did not show a significant change. However, Q-Alb increased over time if the time interval was > 1 year between the measurements. No significant associations between Q-Alb and age, Mini-Mental State Examination, or AD biomarkers were found. Conclusion The increase in Q-Alb suggests that there is an increased leakage through the BBB, which may become more prominent as the disease progresses. This may be a sign of progressive underlying vascular pathology, even in patients with AD without major vascular lesions. More studies are needed to further understand the role of BBB integrity in patients with AD over time and the association with the progression of the disease.
Collapse
Affiliation(s)
- Christian Sandøe Musaeus
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | - Helena Sophia Gleerup
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gunhild Waldemar
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anja Hviid Simonsen
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
19
|
Hillmer L, Erhardt EB, Caprihan A, Adair JC, Knoefel JE, Prestopnik J, Thompson J, Hobson S, Rosenberg GA. Blood-brain barrier disruption measured by albumin index correlates with inflammatory fluid biomarkers. J Cereb Blood Flow Metab 2023; 43:712-721. [PMID: 36522849 PMCID: PMC10108191 DOI: 10.1177/0271678x221146127] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022]
Abstract
Blood-brain barrier (BBB) permeability can be measured by the ratio of albumin in cerebrospinal fluid (CSF) and blood and by dynamic contrast-enhanced MRI (DCEMRI). Albumin is a large molecule measured in CSF and blood to form the albumin index (Qalb), which is a global measure of BBB permeability, while the smaller Gadolinium molecule measures regional transfer (Ktrans); few studies have directly compared them in the same patients. We used both methods as part of a study of mechanisms of white matter injury in patients with different forms of dementia. In addition, we also measured biomarkers for inflammation, including proteases, angiogenic growth factors, and cytokines, and correlated them with the BBB results. We found that there was no correlation between Qalb and Ktrans. The Qalb was associated with the matrix metalloproteinases (MMP-2, MMP-3, and MMP-10), the angiogenic factors (VEGF-C and PlGF), and the cytokines (IL-6, IL-8 and TNF-α). On the other hand, Ktrans was associated with the diffusion measures, mean free water and PSMD, which indicate white matter injury. Our results show that the Qalb and Ktrans measure different aspects of BBB permeability, with albumin being a measure of inflammatory BBB opening and Ktrans indicating white matter injury.
Collapse
Affiliation(s)
- Laura Hillmer
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
| | - Erik B Erhardt
- Department of Mathematics and
Statistics, University of New Mexico, Albuquerque, New Mexico
| | | | - John C Adair
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
- Department of Neurology, University
of New Mexico, Albuquerque, New Mexico
| | - Janice E Knoefel
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
- Department of Neurology, University
of New Mexico, Albuquerque, New Mexico
| | - Jill Prestopnik
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
| | - Jeffrey Thompson
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
| | - Sasha Hobson
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
| | - Gary A Rosenberg
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
- Department of Neurology, University
of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
20
|
Novakova Martinkova J, Ferretti MT, Ferrari A, Lerch O, Matuskova V, Secnik J, Hort J. Longitudinal progression of choroid plexus enlargement is associated with female sex, cognitive decline and ApoE E4 homozygote status. Front Psychiatry 2023; 14:1039239. [PMID: 36970283 PMCID: PMC10031049 DOI: 10.3389/fpsyt.2023.1039239] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/27/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction Choroid plexus (CP)-related mechanisms have been implicated in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease. In this pilot study, we aimed to elucidate the association between longitudinal changes in CP volume, sex and cognitive impairment. Methods We assessed longitudinal changes in CP volume in a cohort of n = 613 subjects across n = 2,334 datapoints from ADNI 2 and ADNI-GO, belonging to cognitively unimpaired (CN), stable mild cognitive impairment (MCI), clinically diagnosed Alzheimer's disease dementia (AD) or convertor (to either AD or MCI) subgroups. CP volume was automatically segmented and used as a response variable in linear mixed effect models with random intercept clustered by patient identity. Temporal effects of select variables were assessed by interactions and subgroup analyses. Results We found an overall significant increase of CP volume in time (14.92 mm3 per year, 95% confidence interval, CI (11.05, 18.77), p < 0.001). Sex-disaggregated results showed an annual rate of increase 9.48 mm3 in males [95% CI (4.08, 14.87), p < 0.001], and 20.43 mm3 in females [95% CI (14.91, 25.93), p < 0.001], indicating more than double the rate of increase in females, which appeared independent of other temporal variables. The only diagnostic group with a significant CP increase as compared to CN was the convertors group, with an increase of 24.88 mm3/year [95% CI (14, 35.82), p < 0.001]. ApoE exhibited a significant temporal effect, with the E4 homozygote group's CP increasing at more than triple the rate of non-carrier or heterozygote groups [40.72, 95% CI (25.97, 55.46), p < 0.001 vs. 12.52, 95% CI (8.02, 17.02), p < 0.001 for ApoE E4 homozygotes and E4 non-carriers, respectively], and may have modified the diagnostic group relationship. Conclusion Our results contribute to potential mechanisms for sex differences in cognitive impairment with a novel finding of twice the annual choroid plexus enlargement in females and provide putative support for CP-related mechanisms of cognitive deterioration and its relationship to ApoE E4.
Collapse
Affiliation(s)
- Julie Novakova Martinkova
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | | | | | - Ondrej Lerch
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Veronika Matuskova
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Juraj Secnik
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
- Center for Alzheimer Research, Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Jakub Hort
- Cognitive Center, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | | |
Collapse
|
21
|
Cui SS, Jiang QW, Chen SD. Sex difference in biological change and mechanism of Alzheimer’s disease: from macro- to micro-landscape. Ageing Res Rev 2023; 87:101918. [PMID: 36967089 DOI: 10.1016/j.arr.2023.101918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 02/16/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and numerous studies reported a higher prevalence and incidence of AD among women. Although women have longer lifetime, longevity does not wholly explain the higher frequency and lifetime risk in women. It is important to understand sex differences in AD pathophysiology and pathogenesis, which could provide foundation for future clinical AD research. Here, we reviewed the most recent and relevant literature on sex differences in biological change of AD from macroscopical neuroimaging to microscopical pathologic change (neuronal degeneration, synaptic dysfunction, amyloid-beta and tau accumulation). We also discussed sex differences in cellular mechanisms related to AD (neuroinflammation, mitochondria dysfunction, oxygen stress, apoptosis, autophagy, blood-brain-barrier dysfunction, gut microbiome alteration, bulk and single cell/nucleus omics) and possible causes underlying these differences including sex-chromosome, sex hormone and hypothalamic-pituitary- adrenal (HPA) axis effects.
Collapse
Affiliation(s)
- Shi-Shuang Cui
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Geriatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian-Wen Jiang
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Geriatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sheng-Di Chen
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
22
|
Relationship between cerebrospinal fluid/serum albumin quotient and phenotype in amyotrophic lateral sclerosis: a retrospective study on 328 patients. Neurol Sci 2023; 44:1679-1685. [PMID: 36646859 DOI: 10.1007/s10072-023-06604-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023]
Abstract
BACKGROUND We analysed the relationship between cerebrospinal fluid (CSF)/serum albumin quotient (Q-Alb) and phenotype in a large cohort of patients with amyotrophic lateral sclerosis (ALS). METHODS Three hundred twenty-eight single-centre consecutive patients with ALS were evaluated for Q-Alb, basic epidemiological and clinical data, motor phenotype, cognitive/behavioural impairment, clinical staging, clinical and neurophysiological indexes of upper (UMN) and lower motor neuron (LMN) dysfunction, and presence of ALS gene mutations. RESULTS Q-Alb did not correlate with age but was independently associated with sex, with male patients having higher levels than female ones; the site of onset was not independently associated with Q-Alb. Q-Alb was not associated with motor phenotype, cognitive/behavioural impairment, disease stage, progression rate, survival, or genetic mutations. Among measures of UMN and LMN dysfunction, Q-Alb only had a weak positive correlation with an electromyography-based index of active limb denervation. CONCLUSION Previous work has documented increased Q-Alb in ALS compared to unaffected individuals. This, together with the absence of associations with nearly all ALS phenotypic features in our cohort, suggests dysfunction of the blood-CSF barrier as a shared, phenotype-independent element in ALS pathophysiology. However, correlation with the active denervation index could point to barrier dysfunction as a local driver of LMN degeneration.
Collapse
|
23
|
Shi Z, Luo K, Deol S, Tan S. A systematic review of noninflammatory cerebrospinal fluid biomarkers for clinical outcome in neonates with perinatal hypoxic brain injury that could be biologically significant. J Neurosci Res 2022; 100:2154-2173. [PMID: 33543500 PMCID: PMC9249405 DOI: 10.1002/jnr.24801] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 01/07/2023]
Abstract
Neonatal encephalopathy (NE) that purportedly arises from hypoxia-ischemia is labeled hypoxic-ischemic encephalopathy (HIE). Perinatal asphyxia is a clinical syndrome involving acidosis, a low Apgar score and the need for resuscitation in the delivery room; asphyxia alerts one to the possibility of NE. In the present systematic review, we focused on the noninflammatory biomarkers in cerebrospinal fluid (CSF) that are involved in the development of possible brain injury in asphyxia or HIE. A literature search in PubMed and EMBASE for case-control studies was conducted and 17 studies were found suitable by a priori criteria. Statistical analysis used the Mantel-Haenszel model for dichotomous data. The pooled mean difference and 95% confidence intervals (CIs) were determined. We identified the best biomarkers, based on the estimation approach in evaluating the biological significance, out of hundreds in three categories: cell adhesion and proliferation, oxidants and antioxidants, and cell damage. The following subtotal-population comparisons were made: perinatal asphyxia versus no asphyxia, asphyxia with HIE versus asphyxia without HIE, asphyxia with HIE versus no asphyxia, and term versus preterm HIE newborn with asphyxia. Biological significance of the biomarkers was determined by using a modification of the estimation approach, by ranking the biomarkers according to the difference in the bounds of the CIs. The most promising CSF biomarkers for prognostication especially for the severest HIE include creatine kinase, xanthine oxidase, vascular endothelial growth factor, neuron-specific enolase, superoxide dismutase, and malondialdehyde. Future studies are recommended using such a combined test to prognosticate the most severely affected patients.
Collapse
Affiliation(s)
- Zhongjie Shi
- Department of Pediatrics, Wayne State University, Detroit, MI, USA
| | - Kehuan Luo
- Department of Pediatrics, Wayne State University, Detroit, MI, USA
| | - Saihaj Deol
- Department of Psychology, College of Liberal Arts & Sciences, Wayne State University, Detroit, MI, USA
| | - Sidhartha Tan
- Department of Pediatrics, Wayne State University, Detroit, MI, USA
| |
Collapse
|
24
|
Travier L, Singh R, Sáenz Fernández D, Deczkowska A. Microbial and immune factors regulate brain maintenance and aging. Curr Opin Neurobiol 2022; 76:102607. [PMID: 35914431 DOI: 10.1016/j.conb.2022.102607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 12/20/2022]
Abstract
Tissue aging can be viewed as a loss of normal maintenance; in advanced age, the mechanisms which keep the tissue healthy on daily bases fail to manage the accumulating "wear and tear", leading to gradual loss of function. In the brain, maintenance is provided primarily by three components: the blood-brain barrier, which allows the influx of certain molecules into the brain while excluding others, the circulation of the cerebrospinal fluid, and the phagocytic function of microglia. Indeed, failure of these systems is associated with cognitive loss and other hallmarks of brain aging. Interestingly, all three mechanisms are regulated not only by internal conditions within the aging brain, but remain highly sensitive to the peripheral signals, such as cytokines or microbiome-derived molecules, present in the systemic circulation. In this article, we discuss the contribution of such peripheral factors to brain maintenance and its loss in aging.
Collapse
Affiliation(s)
- Laetitia Travier
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, F-75015, Paris, France
| | - Roshani Singh
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, F-75015, Paris, France
| | - Daniel Sáenz Fernández
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, F-75015, Paris, France; Universitat de Barcelona, S-08193, Barcelona, Spain
| | - Aleksandra Deczkowska
- Brain-Immune Communication Lab, Institut Pasteur, Université Paris Cité, Inserm U1224, F-75015, Paris, France.
| |
Collapse
|
25
|
Trentini A, Manfrinato MC, Castellazzi M, Bellini T. Sex-Related Differences of Matrix Metalloproteinases (MMPs): New Perspectives for These Biomarkers in Cardiovascular and Neurological Diseases. J Pers Med 2022; 12:jpm12081196. [PMID: 35893290 PMCID: PMC9331234 DOI: 10.3390/jpm12081196] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
It is now established that sex differences occur in clinical manifestation, disease progression, and prognosis for both cardiovascular (CVDs) and central nervous system (CNS) disorders. As such, a great deal of effort is now being put into understanding these differences and turning them into “advantages”: (a) for the discovery of new sex-specific biomarkers and (b) through a review of old biomarkers from the perspective of the “newly” discovered sex/gender medicine. This is also true for matrix metalloproteinases (MMPs), enzymes involved in extracellular matrix (ECM) remodelling, which play a role in both CVDs and CNS disorders. However, most of the studies conducted up to now relegated sex to a mere confounding variable used for statistical model correction rather than a determining factor that can influence MMP levels and, in turn, disease prognosis. Consistently, this approach causes a loss of information that might help clinicians in identifying novel patterns and improve the applicability of MMPs in clinical practice by providing sex-specific threshold values. In this scenario, the current review aims to gather the available knowledge on sex-related differences in MMPs levels in CVDs and CNS conditions, hoping to shed light on their use as sex-specific biomarkers of disease prognosis or progression.
Collapse
Affiliation(s)
- Alessandro Trentini
- Department of Environmental and Prevention Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy;
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Maria Cristina Manfrinato
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (M.C.M.); (T.B.)
| | - Massimiliano Castellazzi
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (M.C.M.); (T.B.)
- Interdepartmental Research Center for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, 44121 Ferrara, Italy
- Correspondence:
| | - Tiziana Bellini
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (M.C.M.); (T.B.)
| |
Collapse
|
26
|
Djukic M, Lange P, Erbguth F, Nau R. Spatial and temporal variation of routine parameters: pitfalls in the cerebrospinal fluid analysis in central nervous system infections. J Neuroinflammation 2022; 19:174. [PMID: 35794632 PMCID: PMC9258096 DOI: 10.1186/s12974-022-02538-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 05/27/2022] [Indexed: 11/10/2022] Open
Abstract
The cerebrospinal fluid (CSF) space is convoluted. CSF flow oscillates with a net flow from the ventricles towards the cerebral and spinal subarachnoid space. This flow is influenced by heartbeats, breath, head or body movements as well as the activity of the ciliated epithelium of the plexus and ventricular ependyma. The shape of the CSF space and the CSF flow preclude rapid equilibration of cells, proteins and smaller compounds between the different parts of the compartment. In this review including reinterpretation of previously published data we illustrate, how anatomical and (patho)physiological conditions can influence routine CSF analysis. Equilibration of the components of the CSF depends on the size of the molecule or particle, e.g., lactate is distributed in the CSF more homogeneously than proteins or cells. The concentrations of blood-derived compounds usually increase from the ventricles to the lumbar CSF space, whereas the concentrations of brain-derived compounds usually decrease. Under special conditions, in particular when distribution is impaired, the rostro-caudal gradient of blood-derived compounds can be reversed. In the last century, several researchers attempted to define typical CSF findings for the diagnosis of several inflammatory diseases based on routine parameters. Because of the high spatial and temporal variations, findings considered typical of certain CNS diseases often are absent in parts of or even in the entire CSF compartment. In CNS infections, identification of the pathogen by culture, antigen detection or molecular methods is essential for diagnosis.
Collapse
|
27
|
Hou M, Zhang B, Fu S, Cai Y, Shi Y. Penetration of Organophosphate Triesters and Diesters across the Blood-Cerebrospinal Fluid Barrier: Efficiencies, Impact Factors, and Mechanisms. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:8221-8230. [PMID: 35658413 DOI: 10.1021/acs.est.2c01850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The penetration of organophosphate triesters (tri-OPEs) and diesters (di-OPEs) across the blood-brain barrier and their influencing factors remain unclear in humans. In this study, 21 tri-OPEs and 8 di-OPEs were measured in 288 paired serum and cerebrospinal fluid (CSF) samples collected in Jinan, China. Six tri-OPEs were frequently detected in both serum and CSF, with median concentrations ranging from 0.062 to 1.62 and 0.042-1.11 ng/mL, respectively. Their penetration efficiencies across the blood-CSF barrier (BCSFB) (RCSF/serum, CCSF/Cserum) were calculated at 0.667-2.80, and these efficiencies first increased and then decreased with their log Kow values. The reduced penetration efficiencies of triphenyl phosphate (TPHP) and 2-ethylhexyl diphenyl phosphate (EHDPP) may be attributed to their strong binding affinities for human serum albumin and p-glycoprotein due to their high hydrophobicity and aryl structure, as indicated by molecular docking. This suggests that active efflux transport may be involved in the penetration of TPHP and EHDPP in addition to passive diffusion similar to the other four tri-OPEs. Di-OPEs were found in few serum samples and even fewer CSF samples, indicating their limited BCSFB permeability. This may be due to their high polarity, low hydrophobicity, and ionic state in blood. This study has important implications for understanding the neurotoxicity of tri-OPEs and di-OPEs and the underlying mechanisms.
Collapse
Affiliation(s)
- Minmin Hou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bona Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Shanji Fu
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Yaqi Cai
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Yali Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| |
Collapse
|
28
|
Castellazzi M, Ferri C, Tecilla G, Huss A, Crociani P, Desina G, Barbella G, Piola A, Permunian S, Senel M, Leone M, Tumani H, Pugliatti M. The Sexual Dimorphism in Cerebrospinal Fluid Protein Content Does Not Affect Intrathecal IgG Synthesis in Multiple Sclerosis. J Pers Med 2022; 12:jpm12060977. [PMID: 35743761 PMCID: PMC9224729 DOI: 10.3390/jpm12060977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS) that mainly affects young adults and females more than males. The detection of intrathecal IgG synthesis (IIS) on cerebrospinal fluid (CSF) analysis supports the diagnosis of MS. A sexual dimorphism has recently been described in CSF protein content. (2) Methods: Clinical and laboratory data from 340 MS patients (F = 231, M = 99) and 89 people with clinically isolated syndrome (CIS) (F = 57, M = 32) were retrospectively analyzed to assess the presence of variables affected by sex and age. (3) Results: In MS, the albumin quotient (QAlb), reflecting the blood–CSF barrier (BCSFB) function, was higher in males (5.6 vs. 4.34) and correlated to age with a constant difference between sexes (F = 41.71). In CIS patients, QAlb increased with age only in males (r = 0.3567). Age was positively correlated to disease duration and severity in MS (r = 0.3502, r = 0.2986, respectively). No differences emerged for quantitative and qualitative IIS determinations. (4) Discussion: Although the main difference between males and females concerns the function of BCSFB assessed by QAlb, this sexual dimorphism does not affect the determination of the IIS evaluated both by quantitative and qualitative methods.
Collapse
Affiliation(s)
- Massimiliano Castellazzi
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (G.T.); (G.B.); (A.P.); (S.P.); (M.P.)
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: ; Tel.: +39-0532-236388
| | - Caterina Ferri
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (G.T.); (G.B.); (A.P.); (S.P.); (M.P.)
| | - Ginevra Tecilla
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (G.T.); (G.B.); (A.P.); (S.P.); (M.P.)
| | - André Huss
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (A.H.); (M.S.); (H.T.)
| | - Paola Crociani
- Neurology Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (P.C.); (G.D.); (M.L.)
| | - Gaetano Desina
- Neurology Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (P.C.); (G.D.); (M.L.)
| | - Gianvito Barbella
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (G.T.); (G.B.); (A.P.); (S.P.); (M.P.)
| | - Alice Piola
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (G.T.); (G.B.); (A.P.); (S.P.); (M.P.)
| | - Samantha Permunian
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (G.T.); (G.B.); (A.P.); (S.P.); (M.P.)
| | - Makbule Senel
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (A.H.); (M.S.); (H.T.)
| | - Maurizio Leone
- Neurology Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (P.C.); (G.D.); (M.L.)
| | - Hayrettin Tumani
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (A.H.); (M.S.); (H.T.)
| | - Maura Pugliatti
- Department of Neurosciences and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (G.T.); (G.B.); (A.P.); (S.P.); (M.P.)
- Interdepartmental Research Center for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
29
|
Abstract
Brain disease remains a significant health, social, and economic burden with a high failure rate of translation of therapeutics to the clinic. Nanotherapeutics have represented a promising area of technology investment to improve drug bioavailability and delivery to the brain, with several successes for nanotherapeutic use for central nervous system disease that are currently in the clinic. However, renewed and continued research on the treatment of neurological disorders is critically needed. We explore the challenges of drug delivery to the brain and the ways in which nanotherapeutics can overcome these challenges. We provide a summary and overview of general design principles that can be applied to nanotherapeutics for uptake and penetration in the brain. We next highlight remaining questions that limit the translational potential of nanotherapeutics for application in the clinic. Lastly, we provide recommendations for ongoing preclinical research to improve the overall success of nanotherapeutics against neurological disease. Expected final online publication date for the Annual Review of Chemical and Biomolecular Engineering, Volume 13 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Andrea Joseph
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, Washington, USA;
| |
Collapse
|
30
|
Chelluboina B, Chokkalla AK, Mehta SL, Morris-Blanco KC, Bathula S, Sankar S, Park JS, Vemuganti R. Tenascin-C induction exacerbates post-stroke brain damage. J Cereb Blood Flow Metab 2022; 42:253-263. [PMID: 34689646 PMCID: PMC9122520 DOI: 10.1177/0271678x211056392] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The role of tenascin-C (TNC) in ischemic stroke pathology is not known despite its prognostic association with cerebrovascular diseases. Here, we investigated the effect of TNC knockdown on post-stroke brain damage and its putative mechanism of action in adult mice of both sexes. Male and female C57BL/6 mice were subjected to transient middle cerebral artery occlusion and injected (i.v.) with either TNC siRNA or a negative (non-targeting) siRNA at 5 min after reperfusion. Motor function (beam walk and rotarod tests) was assessed between days 1 and 14 of reperfusion. Infarct volume (T2-MRI), BBB damage (T1-MRI with contrast), and inflammatory markers were measured at 3 days of reperfusion. The TNC siRNA treated cohort showed significantly curtailed post-stroke TNC protein expression, motor dysfunction, infarction, BBB damage, and inflammation compared to the sex-matched negative siRNA treated cohort. These results demonstrate that the induction of TNC during the acute period after stroke might be a mediator of post-ischemic inflammation and secondary brain damage independent of sex.
Collapse
Affiliation(s)
- Bharath Chelluboina
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA
| | - Suresh L Mehta
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | | | | | - Sneha Sankar
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Jin Soo Park
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA.,William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| |
Collapse
|
31
|
Vascular and blood-brain barrier-related changes underlie stress responses and resilience in female mice and depression in human tissue. Nat Commun 2022; 13:164. [PMID: 35013188 PMCID: PMC8748803 DOI: 10.1038/s41467-021-27604-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022] Open
Abstract
Prevalence, symptoms, and treatment of depression suggest that major depressive disorders (MDD) present sex differences. Social stress-induced neurovascular pathology is associated with depressive symptoms in male mice; however, this association is unclear in females. Here, we report that chronic social and subchronic variable stress promotes blood-brain barrier (BBB) alterations in mood-related brain regions of female mice. Targeted disruption of the BBB in the female prefrontal cortex (PFC) induces anxiety- and depression-like behaviours. By comparing the endothelium cell-specific transcriptomic profiling of the mouse male and female PFC, we identify several pathways and genes involved in maladaptive stress responses and resilience to stress. Furthermore, we confirm that the BBB in the PFC of stressed female mice is leaky. Then, we identify circulating vascular biomarkers of chronic stress, such as soluble E-selectin. Similar changes in circulating soluble E-selectin, BBB gene expression and morphology can be found in blood serum and postmortem brain samples from women diagnosed with MDD. Altogether, we propose that BBB dysfunction plays an important role in modulating stress responses in female mice and possibly MDD.
Collapse
|
32
|
Tsiknia AA, Edland SD, Sundermann EE, Reas ET, Brewer JB, Galasko D, Banks SJ. Sex differences in plasma p-tau181 associations with Alzheimer's disease biomarkers, cognitive decline, and clinical progression. Mol Psychiatry 2022; 27:4314-4322. [PMID: 35768637 PMCID: PMC9718670 DOI: 10.1038/s41380-022-01675-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/27/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023]
Abstract
Studies have shown that women on the Alzheimer's disease (AD) continuum have more pathological tau in the brain and cerebrospinal fluid (CSF), than men. Some studies have found that higher levels of tau biomarkers are more strongly associated with clinical AD, cognitive decline and neurodegeneration in women than in men. Despite major developments in the use of plasma tau phosphorylated at threonine 181 (p-tau181) as an AD biomarker, it is unknown whether these sex differences apply to plasma p-tau181. In 1060 Alzheimer's Disease Neuroimaging Initiative (ADNI) participants (47% women, 73.8 ± 7.6 years old), we examined sex differences in plasma p-tau181 levels and their association with other biomarkers, cognitive decline and incident AD. Linear regressions tested for an effect of sex on plasma p-tau181 levels and for plasma p-tau181 × sex interactions on CSF p-tau181, as well as entorhinal cortex tau, cortical amyloid-β (Aβ) deposition, and brain glucose metabolism, quantified using PET imaging. Linear mixed effects models tested for a sex × baseline plasma p-tau181 interaction on change in cognition over time. Finally, Cox models tested for a sex × plasma p-tau181 interaction on the risk of AD dementia in participants who were free of dementia at baseline. Despite similar plasma p-tau181 levels between sexes, women had lower brain glucose metabolism, greater brain Aβ and entorhinal cortex tau deposition, higher CSF p-tau181 and faster cognitive decline in relation to higher baseline plasma p-tau181 levels compared with men. Among Aβ positive, dementia-free participants, women had higher rates of incident AD dementia associated with increasing baseline plasma p-tau181 levels, relative to men. Our results suggest that sex may impact the clinical interpretation of plasma p-tau181 concentrations. If replicated, these findings could have important implications for the use of plasma p-tau181 as an accessible AD biomarker and screening tool for preventive and therapeutic clinical trials.
Collapse
Affiliation(s)
- Amaryllis A. Tsiknia
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - Steven D. Edland
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093 USA ,grid.266100.30000 0001 2107 4242Division of Biostatistics, School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA 92093 USA
| | - Erin E. Sundermann
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093 USA ,grid.410371.00000 0004 0419 2708Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161 USA
| | - Emilie T. Reas
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - James B. Brewer
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - Douglas Galasko
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - Sarah J. Banks
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093 USA ,grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093 USA
| | | |
Collapse
|
33
|
A Comparison of IgG Index and Oligoclonal Band in the Cerebrospinal Fluid for Differentiating between RRMS and NMOSD. Brain Sci 2021; 12:brainsci12010069. [PMID: 35053810 PMCID: PMC8773790 DOI: 10.3390/brainsci12010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 11/16/2022] Open
Abstract
As the oligoclonal band in the cerebrospinal fluid (CSF-OCB) in predicting relapsing-remitting multiple sclerosis (RRMS) is less sensitive in Asian populations than that in westerners, it remains elusive whether the IgG index could serve as an alternative. The purpose of this study was to compare these two methods of differentiating between RRMS and neuromyelitis optica spectrum disorder (NMOSD) in Chinese patients. A total of 171 patients (81 RRMS and 90 NMOSD) were retrospectively recruited, of whom 82 (56 RRMS and 26 NMOSD) received the CSF-OCB testing additionally. When the onset age was ≤38.5 years, IgG index with the threshold of 0.67 had a significant agreement (k = 0.4, p < 0.001) with the diagnosis while CSF-OCB failed to discriminate (k = 0.1, p = 0.578). However, when the onset age was >38.5 years, both IgG index with the threshold of 0.8 and CSF-OCB were moderately consistent with the diagnosis (both k > 0.4, p < 0.05). In total, our optimized algorithm had the sensitivity, specificity, and predictive accuracy of 0.778, slightly outperforming the CSF-OCB model. Accordingly, a combination of the onset age and IgG index could serve as an alternative to CSF-OCB for differentiating between RRMS and NMOSD in Chinese patients.
Collapse
|
34
|
Oviedo-Salcedo T, Wagner E, Campana M, Gagsteiger A, Strube W, Eichhorn P, Louiset ML, Luykx J, de Witte LD, Kahn RS, Benros ME, Falkai P, Hasan A. Cerebrospinal fluid abnormalities in first- and multi-episode schizophrenia-spectrum disorders: impact of clinical and demographical variables. Transl Psychiatry 2021; 11:621. [PMID: 34880213 PMCID: PMC8654913 DOI: 10.1038/s41398-021-01751-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/18/2021] [Accepted: 11/25/2021] [Indexed: 11/10/2022] Open
Abstract
Multiple lines of evidence indicate that immunological and inflammatory alterations contribute at least in a subgroup to the pathophysiology of schizophrenia. In this retrospective chart review, we investigated whether clinical factors contribute to altered cerebrospinal fluid (CSF) findings in schizophrenia-spectrum disorders. Clinical data from electronic medical records of patients with psychotic disorders (ICD-10: F20-F29) who received routine CSF diagnostics at the Department of Psychiatry and Psychotherapy, LMU Munich, Germany, were included. Chi² tests for dichotomous outcomes and independent t tests for continuous outcomes were used to compare differences between groups. A total of 331 patients were included in the analyses (43.2% female and 56.8% male). The mean age was 37.67 years (±15.58). The mean duration of illness was 71.96 months (±102.59). In all, 40% (128/320) were first-episode psychosis (FEP) patients and 60% (192/320) were multi-episode psychosis (MEP) patients. Elevated CSF protein levels were found in 19.8% and elevated CSF/serum albumin ratios (QAlb) in 29.4% of the cases. Pleocytosis was found in 6.1% of patients. MEP patients showed significantly higher mean QAlb compared with FEP patients (t(304.57) = -2.75, p = 0.006), which did not remain significant after correcting for age. QAlb elevation occurred more frequently in men (X2(1) = 14.76, p = <0.001). For treatment resistance, family history, and cMRI alterations, no significant differences in CSF-related outcomes were detected. Our work extends other retrospective cohorts confirming a relevant degree of CSF alterations in schizophrenia-spectrum disorders and shows the difficulty to relate these alterations to clinical and disease course trajectories. More research is needed to develop treatment response predictors from CSF analyses.
Collapse
Affiliation(s)
- Tatiana Oviedo-Salcedo
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Elias Wagner
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.
| | - Mattia Campana
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Anna Gagsteiger
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Strube
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Bezirkskrankenhaus Augsburg, Augsburg, Germany
| | - Peter Eichhorn
- Institute of Laboratory Medicine, Klinikum der Universität München, Ludwig Maximilians-University Munich, Munich, Germany
| | - Marie-Luise Louiset
- Institute of Laboratory Medicine, Klinikum der Universität München, Ludwig Maximilians-University Munich, Munich, Germany
| | - Jurjen Luykx
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University, Medical Center Utrecht, Utrecht, The Netherlands
| | - Lot D de Witte
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - René S Kahn
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael E Benros
- Biological and Precision Psychiatry, Copenhagen Research Centre for Mental Health, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Bezirkskrankenhaus Augsburg, Augsburg, Germany
| |
Collapse
|
35
|
Evidence of Neuroinflammation and Blood-Brain Barrier Disruption in Women with Preeclampsia and Eclampsia. Cells 2021; 10:cells10113045. [PMID: 34831266 PMCID: PMC8616341 DOI: 10.3390/cells10113045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/16/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Cerebral complications in preeclampsia are leading causes of maternal mortality. Animal models suggest that an injured blood-brain barrier and neuroinflammation may be important but there is paucity of data from human studies. Therefore, we aimed to evaluate this in women with preeclampsia and eclampsia. We included women recruited to the South African Preeclampsia Obstetric Adverse Events (PROVE) biobank. Blood and cerebrospinal fluid (CSF) were collected around delivery. CSF was analyzed for neuroinflammatory markers interleukin 1β, interleukin 6, interleukin-8 and tumor necrosis factor alpha (TNF-alpha). The CSF to plasma albumin ratio was measured to assess blood-brain barrier function. Women with eclampsia (n = 4) showed increased CSF concentrations of all pro-inflammatory cytokines and TNF-alpha compared to women with normotensive pregnancies (n = 7) and also for interleukin-6 and TNF-alpha compared to women with preeclampsia (n = 4). Women with preeclampsia also showed increases in pro-inflammatory cytokines IL-6 and IL-8 but not TNF-alpha in the CSF compared to women with normotensive pregnancies. In particular, women with eclampsia but also women with preeclampsia showed an increase in the CSF to plasma albumin ratio compared to normotensive women. In conclusion, women with preeclampsia and eclampsia show evidence of neuroinflammation and an injured blood-brain barrier. These findings are seen in particular among women with eclampsia.
Collapse
|
36
|
Galea I. The blood-brain barrier in systemic infection and inflammation. Cell Mol Immunol 2021; 18:2489-2501. [PMID: 34594000 PMCID: PMC8481764 DOI: 10.1038/s41423-021-00757-x] [Citation(s) in RCA: 225] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/04/2021] [Indexed: 02/08/2023] Open
Abstract
The vascular blood-brain barrier is a highly regulated interface between the blood and brain. Its primary function is to protect central neurons while signaling the presence of systemic inflammation and infection to the brain to enable a protective sickness behavior response. With increasing degrees and duration of systemic inflammation, the vascular blood-brain barrier becomes more permeable to solutes, undergoes an increase in lymphocyte trafficking, and is infiltrated by innate immune cells; endothelial cell damage may occasionally occur. Perturbation of neuronal function results in the clinical features of encephalopathy. Here, the molecular and cellular anatomy of the vascular blood-brain barrier is reviewed, first in a healthy context and second in a systemic inflammatory context. Distinct from the molecular and cellular mediators of the blood-brain barrier's response to inflammation, several moderators influence the direction and magnitude at genetic, system, cellular and molecular levels. These include sex, genetic background, age, pre-existing brain pathology, systemic comorbidity, and gut dysbiosis. Further progress is required to define and measure mediators and moderators of the blood-brain barrier's response to systemic inflammation in order to explain the heterogeneity observed in animal and human studies.
Collapse
Affiliation(s)
- Ian Galea
- grid.5491.90000 0004 1936 9297Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| |
Collapse
|
37
|
Alisch JSR, Kiely M, Triebswetter C, Alsameen MH, Gong Z, Khattar N, Egan JM, Bouhrara M. Characterization of Age-Related Differences in the Human Choroid Plexus Volume, Microstructural Integrity, and Blood Perfusion Using Multiparameter Magnetic Resonance Imaging. Front Aging Neurosci 2021; 13:734992. [PMID: 34603011 PMCID: PMC8485051 DOI: 10.3389/fnagi.2021.734992] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/30/2021] [Indexed: 11/13/2022] Open
Abstract
The choroid plexus (CP) is an important cerebral structure involved in cerebrospinal fluid production and transport of solutes into the brain. Recent studies have uncovered the involvement of the CP in neurological disorders such as Alzheimer's disease and multiple sclerosis. However, our understanding of human age-related microstructural and functional changes in the CP with aging and neuropathology is limited. In this cross-sectional study, we investigated age and sex differences in the CP structure and function using advanced quantitative magnetic resonance imaging methodology in a large cohort (n = 155) of cognitively unimpaired individuals over a wide age range between 21 and 94 years. Our analysis included volumetric measurements, relaxometry measures (T 1 and T 2), diffusion tensor imaging (DTI) measures of fractional anisotropy (FA) and mean diffusivity (MD), as well as measures of cerebral blood flow (CBF). Our results revealed that CP volume was increasing with advancing age. We conjecture that this novel observation is likely attributed to alterations in the CP microstructure or function as well as to ventriculomegaly. Indeed, we also found that CBF was lower with advanced age, while, consistent with previous studies, T 1, T 2 and MD were higher, and FA was lower with advanced age. We attribute these functional and microstructural differences to a deteriorated CP structural integrity with aging. Furthermore, our relaxometry and DTI measures were found to be associated with differences in blood perfusion revealing lower microstructural integrity with lower CBF. Finally, in agreement with literature, sex-related differences in MD and CBF were statistically significant. This work lays the foundation for ongoing investigation of the involvement of CP in neurodegeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mustapha Bouhrara
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
38
|
Castellazzi M, Pizzicotti S, Lombardo I, Alfiero S, Morotti A, Pellegatti P, Negri G, Natali L, Ferri C, Fainardi E, Bellini T, Pugliatti M. Sexual dimorphism in the cerebrospinal fluid total protein content. Clin Chem Lab Med 2021; 58:1885-1890. [PMID: 32598300 DOI: 10.1515/cclm-2020-0419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/18/2020] [Indexed: 11/15/2022]
Abstract
Objectives Cerebrospinal fluid (CSF) is a clear, colorless body fluid filling the central nervous system. The determination of the CSF total protein (TP) content represents an important screening test of various pathologies. We aimed to address the effect of sex and age on CSF-TP content and the use of the current upper reference limits (URLs). Methods CSF-TP content was analysed in a selected population of 1,252 patients (648 women and 604 men; age 18-89 years) who underwent lumbar puncture as a part of the diagnostic work-up. Samples presenting (i) more than 5 white blood cells (WBC)/µL, (ii) discolorations and (iii) reduced glucose were not included. Results The CSF-TP content median values were significantly higher in men than in women (46 vs. 37 mg/dL) even after adjusting for age and different hospital inpatients. CSF-TP content positively correlated with age both in men and in women with a constant difference between sexes of 8.5 mg/dL. Applying the most used URLs (mainly 45 and 50 mg/dL, but also 60 mg/dL), men received a laboratory report suggestive of altered CSF-TP content more frequently than women. The use of age- and sex-calibrated CSF-TP URLs reduced, but not eliminated, this sex-gap. Conclusions Using the current URLs, a condition of "elevated CSF-TP content" may be overestimated in men or, conversely, underestimated in women, regardless of the age and of the diagnosis. These results highlighted the need to apply CSF-TP URLs values normalized for both sex and age.
Collapse
Affiliation(s)
- Massimiliano Castellazzi
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
- Interdepartmental Research Center for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, Ferrara, Italy
| | - Stefano Pizzicotti
- Chemical-Clinical Analysis Laboratory, "S. Anna" University Hospital, Ferrara, Italy
| | - Ilenia Lombardo
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Sarah Alfiero
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Patrizia Pellegatti
- Chemical-Clinical Analysis Laboratory, "S. Anna" University Hospital, Ferrara, Italy
| | - Giovanna Negri
- Chemical-Clinical Analysis Laboratory, "S. Anna" University Hospital, Ferrara, Italy
| | - Lara Natali
- Chemical-Clinical Analysis Laboratory, "S. Anna" University Hospital, Ferrara, Italy
| | - Caterina Ferri
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Enrico Fainardi
- Interdepartmental Research Center for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, Ferrara, Italy
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Tiziana Bellini
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
- University Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Maura Pugliatti
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
- Interdepartmental Research Center for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, Ferrara, Italy
| |
Collapse
|
39
|
Solarz A, Majcher-Maślanka I, Chocyk A. Effects of early-life stress and sex on blood-brain barrier permeability and integrity in juvenile and adult rats. Dev Neurobiol 2021; 81:861-876. [PMID: 34320279 DOI: 10.1002/dneu.22846] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/22/2022]
Abstract
Early-life stress (ELS) is considered a relevant etiological factor for neurodegenerative and mental disorders. In the present study, we hypothesized that ELS may persistently and sex dependently influence blood-brain barrier (BBB) integrity and function during critical periods of brain development and consequently determine susceptibility to and sex-related prevalence of chronic diseases in adult life. We used the maternal separation (MS) procedure in rats to model ELS and evaluated BBB permeability and gene expression of selected tight junction (TJ) proteins, glucose transporter type 1 (Slc2a1) and aquaporin 4 (Aqp4) in the medial prefrontal cortex (mPFC), dorsal striatum (dSTR) and hippocampus of juvenile and adult rats. Serum concentrations of a peripheral marker of BBB function (S100β) and proinflammatory cytokines were also assessed. We observed developmental sealing of the BBB and sex differences in the permeability of the BBB and the mRNA expression of TJ proteins and Slc2a1. Adult females showed lower BBB permeability and higher levels of Cldn3, Cldn5, Ocln, and Slc2a1 in the mPFC and dSTR than males. MS temporarily increased BBB permeability in the dSTR of juvenile males and affected mRNA expression of the majority of studied proteins related to BBB function in age-, region- and sex-dependent manners. Additionally, MS sex dependently decreased serum S100β levels and did not affect proinflammatory cytokine concentrations. In general, our study did not reveal a clear or strong negative effect of MS on BBB integrity. However, the results suggest that ELS may induce adaptive/maladaptive changes or compensatory mechanisms within the BBB of unknown yet consequences.
Collapse
Affiliation(s)
- Anna Solarz
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Agnieszka Chocyk
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
40
|
Castellazzi M, Ferri C, Alfiero S, Lombardo I, Laudisi M, Tecilla G, Boni M, Pizzicotti S, Fainardi E, Bellini T, Pugliatti M. Sex-Related Differences in Cerebrospinal Fluid Plasma-Derived Proteins of Neurological Patients. Diagnostics (Basel) 2021; 11:diagnostics11050884. [PMID: 34065720 PMCID: PMC8156001 DOI: 10.3390/diagnostics11050884] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/03/2021] [Accepted: 05/14/2021] [Indexed: 11/16/2022] Open
Abstract
Background and aims: Cerebrospinal fluid (CSF) protein content presents a sexual dimorphism in humans. We investigated sex-related differences in CSF IgG levels and in the quantification of intrathecal IgG synthesis (IIS). Methods: CSF, serum albumin and IgG were measured in 1519 neurological patients and both linear and hyperbolic formulas were used for the quantification of IIS. CSF-restricted oligoclonal IgG bands (OCBs) were used as “gold standard”. Results: The linear IgG Index showed a weak agreement with OCBs in males and females (k = 0.559, k = 0.587, respectively), while the hyperbolic Reiber’s formulas had a moderate agreement with OCBs in females (k = 0.635) and a weak agreement in males (k = 0.565). Higher CSF albumin and IgG levels were found in men than in women in the whole population and in subjects without IIS after adjusting for age and for serum concentrations of albumin and IgG, respectively (Quade statistics, p < 0.000001). CSF and serum albumin and IgG levels positively correlated to age in both sexes. CSF total protein content did not correlate with CSF leukocyte numbers but was higher in patients with marked pleocytosis. Conclusions: In neurological patients, men have higher levels of CSF serum-derived proteins, such as albumin and IgG.
Collapse
Affiliation(s)
- Massimiliano Castellazzi
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (S.A.); (I.L.); (M.L.); (G.T.); (T.B.); (M.P.)
- Interdepartmental Research Center for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: ; Tel.: +39-0532-236388
| | - Caterina Ferri
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (S.A.); (I.L.); (M.L.); (G.T.); (T.B.); (M.P.)
| | - Sarah Alfiero
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (S.A.); (I.L.); (M.L.); (G.T.); (T.B.); (M.P.)
| | - Ilenia Lombardo
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (S.A.); (I.L.); (M.L.); (G.T.); (T.B.); (M.P.)
| | - Michele Laudisi
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (S.A.); (I.L.); (M.L.); (G.T.); (T.B.); (M.P.)
| | - Ginevra Tecilla
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (S.A.); (I.L.); (M.L.); (G.T.); (T.B.); (M.P.)
| | - Michela Boni
- Chemical-Clinical Analysis Laboratory, “S. Anna” University Hospital, 44124 Ferrara, Italy; (M.B.); (S.P.)
| | - Stefano Pizzicotti
- Chemical-Clinical Analysis Laboratory, “S. Anna” University Hospital, 44124 Ferrara, Italy; (M.B.); (S.P.)
| | - Enrico Fainardi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50121 Florence, Italy;
| | - Tiziana Bellini
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (S.A.); (I.L.); (M.L.); (G.T.); (T.B.); (M.P.)
- Interdepartmental Research Center for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, 44121 Ferrara, Italy
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Maura Pugliatti
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (C.F.); (S.A.); (I.L.); (M.L.); (G.T.); (T.B.); (M.P.)
- Interdepartmental Research Center for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
41
|
Musaeus CS, Gleerup HS, Høgh P, Waldemar G, Hasselbalch SG, Simonsen AH. Cerebrospinal Fluid/Plasma Albumin Ratio as a Biomarker for Blood-Brain Barrier Impairment Across Neurodegenerative Dementias. J Alzheimers Dis 2021; 75:429-436. [PMID: 32280104 DOI: 10.3233/jad-200168] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previous studies have shown an association between disruption of the blood-brain-barrier (BBB) and dementias of different etiologies. The protein concentration of cerebrospinal fluid (CSF) can be used as an indirect measurement for the permeability of the BBB using the CSF/plasma albumin quotient (Q-Alb) or total CSF protein. OBJECTIVE In the current study, we wanted to investigate Q-Alb and CSF protein concentration in dementias of different etiologies and the possible confounding factors. METHODS A total of 510 patients and healthy controls were included in the current study. The patients were diagnosed with Alzheimer's disease (AD), dementia with Lewy bodies (DLB), vascular dementia (VaD), or frontotemporal dementia (FTD). RESULTS We found that Q-Alb was significantly different between the groups (p = 0.002, F = 3.874). Patients with DLB and VaD showed the largest Q-Alb. Although not significant for CSF total protein, we found the same overall pattern for DLB and VaD. When examining confounding factors, we found a positive association with age and a lower Fazekas score in DLB as compared to VaD. CONCLUSION These results suggest that Q-Alb can contribute to our understanding of the pathophysiological mechanisms in DLB, and Q-Alb may serve as a supplementary diagnostic marker. Furthermore, we found a positive association with age, which may be due to differences in vascular co-morbidities. In addition, in patients with DLB, the increased Q-Alb is not due to vascular lesions. Studies are needed to validate the possible diagnostic value of Q-Alb in a larger cohort.
Collapse
Affiliation(s)
- Christian Sandøe Musaeus
- Department of Neurology, Danish Dementia Research Centre (DDRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Helena Sophia Gleerup
- Department of Neurology, Danish Dementia Research Centre (DDRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Peter Høgh
- Regional Dementia Research Centre, Department of Neurology, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gunhild Waldemar
- Department of Neurology, Danish Dementia Research Centre (DDRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Department of Neurology, Danish Dementia Research Centre (DDRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anja Hviid Simonsen
- Department of Neurology, Danish Dementia Research Centre (DDRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
McKetney J, Panyard DJ, Johnson SC, Carlsson CM, Engelman CD, Coon JJ. Pilot proteomic analysis of cerebrospinal fluid in Alzheimer's disease. Proteomics Clin Appl 2021; 15:e2000072. [PMID: 33682374 PMCID: PMC8197734 DOI: 10.1002/prca.202000072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 01/10/2023]
Abstract
Proteomic analysis of cerebrospinal fluid (CSF) holds great promise in understanding the progression of neurodegenerative diseases, including Alzheimer's disease (AD). As one of the primary reservoirs of neuronal biomolecules, CSF provides a window into the biochemical and cellular aspects of the neurological environment. CSF can be drawn from living participants allowing the potential alignment of clinical changes with these biochemical markers. Using cutting-edge mass spectrometry technologies, we perform a streamlined proteomic analysis of CSF. We quantify greater than 700 proteins across 10 pairs of age- and sex-matched participants in approximately one hour of analysis time each. Using the paired participant study structure, we identify a small group of biologically relevant proteins that show substantial changes in abundance between cognitive normal and AD participants, which were then analyzed at the peptide level using parallel reaction monitoring experiments. Our findings suggest the utility of fractionating a single sample and using matching to increase proteomic depth in cerebrospinal fluid, as well as the potential power of an expanded study.
Collapse
Affiliation(s)
- Justin McKetney
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI
- National Center for Quantitative Biology of Complex Systems, Madison, WI
| | - Daniel J. Panyard
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI
| | - Sterling C. Johnson
- Geriatric Research Education and Clinical Center, Middleton Memorial Veterans Hospital, Madison, WI
- Wisconsin Alzheimer’s Institute, University of Wisconsin-Madison School of Medicine, Madison, WI
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine, Madison, WI
| | - Cynthia M. Carlsson
- Geriatric Research Education and Clinical Center, Middleton Memorial Veterans Hospital, Madison, WI
- Wisconsin Alzheimer’s Institute, University of Wisconsin-Madison School of Medicine, Madison, WI
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine, Madison, WI
| | - Corinne D. Engelman
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI
- Wisconsin Alzheimer’s Institute, University of Wisconsin-Madison School of Medicine, Madison, WI
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine, Madison, WI
| | - Joshua J. Coon
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI
- National Center for Quantitative Biology of Complex Systems, Madison, WI
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI
- Morgridge Institute for Research, Madison, WI
| |
Collapse
|
43
|
Mielke MM. Consideration of Sex Differences in the Measurement and Interpretation of Alzheimer Disease-Related Biofluid-Based Biomarkers. J Appl Lab Med 2021; 5:158-169. [PMID: 31811073 DOI: 10.1373/jalm.2019.030023] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND The development of cerebrospinal fluid and blood-based biomarkers for Alzheimer disease (AD) and related disorders is rapidly progressing. Such biomarkers may be used clinically to screen the population, to enhance diagnosis, or to help determine prognosis. Although the use of precision medicine methods has contributed to enhanced understanding of the AD pathophysiological changes and development of assays, one aspect not commonly considered is sex differences. CONTENT There are several ways in which sex can affect the concentration or interpretation of biofluid biomarkers. For some markers, concentrations will vary by sex. For others, the concentrations might not vary by sex, but the impact or interpretation may vary by sex depending on the context of use (e.g., diagnostic vs prognostic). Finally, for others, there will be no sex differences in concentrations or their interpretation. This review will first provide a basis for sex differences, including differences in brain structure and function, and the means by which these differences could contribute to sex differences in biomarker concentrations. Next, the current state of sex differences in AD-related biofluid markers (i.e., amyloid-β, phosphorylated τ, total τ, neurofilament light chain, and neurogranin) will be reviewed. Lastly, factors that can lead to the misinterpretation of observed sex differences in biomarkers (either providing evidence for or against) will be considered. SUMMARY This review is intended to provide an impetus to consider sex differences in the measurement and interpretation of AD-related biofluid-based biomarkers.
Collapse
Affiliation(s)
- Michelle M Mielke
- Departments of Health Sciences Research and Neurology, Mayo Clinic, Rochester, MN
| |
Collapse
|
44
|
Skillbäck T, Blennow K, Zetterberg H, Shams S, Machado A, Pereira J, Lindberg O, Mielke MM, Zettergren A, Ryden L, Westman E, Wahlund L, Skoog I, Kern S. Sex differences in CSF biomarkers for neurodegeneration and blood-brain barrier integrity. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12141. [PMID: 33748393 PMCID: PMC7968119 DOI: 10.1002/dad2.12141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/21/2020] [Accepted: 12/02/2020] [Indexed: 11/18/2022]
Abstract
INTRODUCTION As cerebrospinal fluid (CSF) neurofilament light protein (NfL) and the CSF/serum albumin ratio (QAlb) are used in the clinical routine, the impact of demographic factors on these biomarkers is important to understand. METHODS Participants were derived from two Swedish samples: the population-based H70 Study (n = 308, age 70) and a clinical routine cohort (CSF NfL, n = 8995, QAlb, n = 39252, age 0 to 95). In the population-based study, QAlb and NfL were examined in relation to sex, cardiovascular risk factors, and cerebral white matter lesions (WMLs). In the clinical cohort, QAlb and NfL sex differences were tested in relation to age. RESULTS Men had higher QAlb and NfL concentrations and had higher QAlb and NfL concentrations from adolescence throughout life. NfL was not related to WML, but QAlb correlated positively with WMLs. DISCUSSION The CSF NfL sex difference could not be explained by vascular pathology. Future studies should consider using different reference limits for men and women.
Collapse
Affiliation(s)
- Tobias Skillbäck
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Sara Shams
- Department of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Alejandra Machado
- Department of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Joana Pereira
- Department of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Olof Lindberg
- Department of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Michelle M. Mielke
- Department of Health Sciences ResearchDivision of Epidemiology and Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Anna Zettergren
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgSweden
| | - Lina Ryden
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgSweden
| | - Eric Westman
- Department of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
- Department of NeuroimagingCentre for Neuroimaging SciencesInstitute of PsychiatryPsychology and Neuroscience, King's College LondonLondonUnited Kingdom
| | - Lars‐Olof Wahlund
- Department of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Ingmar Skoog
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgSweden
| | - Silke Kern
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgSweden
| |
Collapse
|
45
|
Abstract
The blood-brain barrier (BBB) protects the central nervous system (CNS) from unregulated exposure to the blood and its contents. The BBB also controls the blood-to-brain and brain-to-blood permeation of many substances, resulting in nourishment of the CNS, its homeostatic regulation and communication between the CNS and peripheral tissues. The cells forming the BBB communicate with cells of the brain and in the periphery. This highly regulated interface changes with healthy aging. Here, we review those changes, starting with morphology and disruption. Transporter changes include those for amyloid beta peptide, glucose and drugs. Brain fluid dynamics, pericyte health and basement membrane and glycocalyx compositions are all altered with healthy aging. Carrying the ApoE4 allele leads to an acceleration of most of the BBB's age-related changes. We discuss how alterations in the BBB that occur with healthy aging reflect adaptation to the postreproductive phase of life and may affect vulnerability to age-associated diseases.
Collapse
|
46
|
Sex-Related Differences in Regional Blood-Brain Barrier Integrity in Non-Demented Elderly Subjects. Int J Mol Sci 2021; 22:ijms22062860. [PMID: 33799794 PMCID: PMC8001339 DOI: 10.3390/ijms22062860] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 01/01/2023] Open
Abstract
The role of the blood-brain barrier (BBB) breakdown has been recognized as being important in Alzheimer's disease pathogenesis. We aimed to evaluate whether regional BBB integrity differed according to sex and whether differences in BBB integrity changed as a consequence of aging or cognitive decline, using dynamic contrast-enhanced (DCE)-magnetic resonance imaging (MRI). In total, 75 participants with normal cognition (NC) or mild cognitive impairment (MCI) underwent cognitive assessments and MRI examination including DCE-MRI. Regional Ktrans was calculated in cortical regions and the Patlak permeability model was used to calculate BBB permeability (Ktrans, min-1). Females had a lower median Ktrans in the cingulate and occipital cortices. In the "older old" group, sex differences in Ktrans were only observed in the occipital cortex. In the MCI group, sex differences in Ktrans were only observed in the occipital cortex. Age was the only predictor of cognitive assessment scores in the male MCI group; however, educational years and Ktrans in the occipital cortex could predict cognitive scores in the female MCI group. Our study revealed that females may have better BBB integrity in cingulate and occipital cortices. We also found that sex-related differences in BBB integrity are attenuated with aging or cognitive decline.
Collapse
|
47
|
Sandebring-Matton A, Goikolea J, Björkhem I, Paternain L, Kemppainen N, Laatikainen T, Ngandu T, Rinne J, Soininen H, Cedazo-Minguez A, Solomon A, Kivipelto M. 27-Hydroxycholesterol, cognition, and brain imaging markers in the FINGER randomized controlled trial. Alzheimers Res Ther 2021; 13:56. [PMID: 33676572 PMCID: PMC7937194 DOI: 10.1186/s13195-021-00790-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/15/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND 27-Hydroxycholesterol (27-OH), the main circulating oxysterol in humans and the potential missing link between peripheral hypercholesterolemia and Alzheimer's disease (AD), has not been investigated previously in relation to cognition and neuroimaging markers in the context of preventive interventions. METHODS The 2-year Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) included older individuals (60-77 years) at increased risk for dementia but without dementia or substantial cognitive impairment from the general population. Participants were randomized to a multidomain intervention (diet, exercise, cognitive training, and vascular risk management) or control group (general health advice) in a 1:1 ratio. Outcome assessors were masked to group allocation. This FINGER exploratory sub-study included 47 participants with measures of 27-OH, cognition, brain MRI, brain FDG-PET, and PiB-PET. Linear regression models were used to assess the cross-sectional and longitudinal associations between 27-OH, cognition, and neuroimaging markers, considering several potential confounders/intervention effect modifiers. RESULTS 27-OH reduction during the intervention was associated with improvement in cognition (especially memory). This was not observed in the control group. The intervention reduced 27-OH particularly in individuals with the highest 27-OH levels and younger age. No associations were found between changes in 27-OH levels and neuroimaging markers. However, at baseline, a higher 27-OH was associated with lower total gray matter and hippocampal volume, and lower cognitive scores. These associations were unaffected by total cholesterol levels. While sex seemed to influence associations at baseline, it did not affect longitudinal associations. CONCLUSION 27-OH appears to be a marker not only for dementia/AD risk, but also for monitoring the effects of preventive interventions on cholesterol metabolism. TRIAL REGISTRATION ClinicalTrials.gov , NCT01041989 . Registered on 4 January 2010.
Collapse
Affiliation(s)
- Anna Sandebring-Matton
- Division of Neurogeriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden.
- Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden.
| | - Julen Goikolea
- Division of Neurogeriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
| | - Ingemar Björkhem
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Laura Paternain
- Division of Neurogeriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
| | - Nina Kemppainen
- Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Tiina Laatikainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Joint Municipal Authority for North Karelia Social and Health Services, Joensuu, Finland
- Public Health Promotion Unit, Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Tiia Ngandu
- Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
- Public Health Promotion Unit, Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Juha Rinne
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
- Neurocenter, Neurology Kuopio University Hospital, Kuopio, Finland
| | - Angel Cedazo-Minguez
- Division of Neurogeriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
| | - Alina Solomon
- Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
- Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
48
|
Parrado-Fernandez C, Leoni V, Saeed A, Rodriguez-Rodriguez P, Sandebring-Matton A, Córdoba-Beldad CM, Bueno P, Gali CC, Panzenboeck U, Cedazo-Minguez A, Björkhem I. Sex difference in flux of 27-hydroxycholesterol into the brain. Br J Pharmacol 2021; 178:3194-3204. [PMID: 33345295 PMCID: PMC8359195 DOI: 10.1111/bph.15353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 01/21/2023] Open
Abstract
Background and Purpose The cerebrospinal fluid (CSF)/plasma albumin ratio (QAlb) is believed to reflect the integrity of the blood–brain barrier (BBB). Recently, we reported that QAlb is lower in females. This may be important for uptake of neurotoxic 27‐hydroxycholesterol (27OH) by the brain in particular because plasma levels of 27OH are higher in males. We studied sex differences in the relation between CSF and plasma levels of 27OH and its major metabolite 7α‐hydroxy‐3‐oxo‐4‐cholestenoic acid (7HOCA) with QAlb. We tested the possibility of sex differences in the brain metabolism of 27OH and if its flux into the brain disrupted integrity of the BBB. Experimental Approach We have examined our earlier studies looking for sex differences in CSF levels of oxysterols and their relation to QAlb. We utilized an in vitro model for the BBB with primary cultured brain endothelial cells to test if 27OH has a disruptive effect on this barrier. We measured mRNA and protein levels of CYP7B1 in autopsy brain samples. Key Results The correlation between CSF levels of 27OH and QAlb was higher in males whereas, with 7HOCA, the correlation was higher in females. No significant sex difference in the expression of CYP7B1 mRNA in brain autopsy samples. A correlation was found between plasma levels of 27OH and QAlb. No support was obtained for the hypothesis that plasma levels of 27OH have a disruptive effect on the BBB. Conclusions and Implications The sex differences are discussed in relation to negative effects of 27OH on different brain functions. LINKED ARTICLES This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc
Collapse
Affiliation(s)
- Cristina Parrado-Fernandez
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden.,Discovery and Research, AlzeCure Pharma AB, Huddinge, Sweden
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Desio, ASST-Monza and School of Medicine, University of Milano Bicocca, Monza, Italy
| | - Ahmed Saeed
- Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | | | - Anna Sandebring-Matton
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden
| | | | - Paula Bueno
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden
| | - Chaitanya Chakravarthi Gali
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Ute Panzenboeck
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Angel Cedazo-Minguez
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden.,Rare & Neurologic Diseases Research Therapeutic Area, Neurodegeneration Research, Sanofi Pharmaceuticals, Paris, France
| | - Ingemar Björkhem
- Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| |
Collapse
|
49
|
Siniscalchi A, Sztajzel R, Murphy S, De Sarro G, Gallelli L. Blood-brain barrier dysfunction and extension of time window for thrombolysis: a limitation not evaluated. Curr Vasc Pharmacol 2021; 19:585-586. [PMID: 33535958 DOI: 10.2174/1570161119666210203144947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/09/2021] [Accepted: 01/14/2021] [Indexed: 11/22/2022]
Affiliation(s)
- Antonio Siniscalchi
- Department of Neurology and Stroke Unit, Annunziata Hospital of Cosenza, Cosenza. Italy
| | - Roman Sztajzel
- Department of Neurology, University Hospital of Geneva, Geneva. Switzerland
| | - Sean Murphy
- General Medicine, Stroke Unit, Mater Misericordiae University Hospital, Dublin. Ireland
| | - Giovambattista De Sarro
- University of Catanzaro, Clinical Pharmacology Unit, Mater Domini University Hospital - Chair of Pharmacology, Department of Health Science, School of Medicine, Catanzaro. Italy
| | - Luca Gallelli
- University of Catanzaro, Clinical Pharmacology Unit, Mater Domini University Hospital - Chair of Pharmacology, Department of Health Science, School of Medicine, Catanzaro. Italy
| |
Collapse
|
50
|
Dudek KA, Dion‐Albert L, Kaufmann FN, Tuck E, Lebel M, Menard C. Neurobiology of resilience in depression: immune and vascular insights from human and animal studies. Eur J Neurosci 2021; 53:183-221. [PMID: 31421056 PMCID: PMC7891571 DOI: 10.1111/ejn.14547] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
Major depressive disorder (MDD) is a chronic and recurrent psychiatric condition characterized by depressed mood, social isolation and anhedonia. It will affect 20% of individuals with considerable economic impacts. Unfortunately, 30-50% of depressed individuals are resistant to current antidepressant treatments. MDD is twice as prevalent in women and associated symptoms are different. Depression's main environmental risk factor is chronic stress, and women report higher levels of stress in daily life. However, not every stressed individual becomes depressed, highlighting the need to identify biological determinants of stress vulnerability but also resilience. Based on a reverse translational approach, rodent models of depression were developed to study the mechanisms underlying susceptibility vs resilience. Indeed, a subpopulation of animals can display coping mechanisms and a set of biological alterations leading to stress resilience. The aetiology of MDD is multifactorial and involves several physiological systems. Exacerbation of endocrine and immune responses from both innate and adaptive systems are observed in depressed individuals and mice exhibiting depression-like behaviours. Increasing attention has been given to neurovascular health since higher prevalence of cardiovascular diseases is found in MDD patients and inflammatory conditions are associated with depression, treatment resistance and relapse. Here, we provide an overview of endocrine, immune and vascular changes associated with stress vulnerability vs. resilience in rodents and when available, in humans. Lack of treatment efficacy suggests that neuron-centric treatments do not address important causal biological factors and better understanding of stress-induced adaptations, including sex differences, could contribute to develop novel therapeutic strategies including personalized medicine approaches.
Collapse
Affiliation(s)
- Katarzyna A. Dudek
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Laurence Dion‐Albert
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Fernanda Neutzling Kaufmann
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Ellen Tuck
- Smurfit Institute of GeneticsTrinity CollegeDublinIreland
| | - Manon Lebel
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Caroline Menard
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| |
Collapse
|