1
|
Hidaka Y, Hashimoto M, Suehiro T, Fukuhara R, Ishikawa T, Tsunoda N, Koyama A, Honda K, Miyagawa Y, Yoshiura K, Yuuki S, Kajitani N, Boku S, Ishii K, Ikeda M, Takebayashi M. Association between choroid plexus volume and cognitive function in community-dwelling older adults without dementia: a population-based cross-sectional analysis. Fluids Barriers CNS 2024; 21:101. [PMID: 39696504 DOI: 10.1186/s12987-024-00601-0] [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: 08/15/2024] [Accepted: 11/16/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND An increase in choroid plexus (CP) volume may be associated with cognitive decline in older individuals without dementia. In this study, we aimed to clarify whether CP volume can serve as an imaging marker of cognitive decline, determine how strongly CP volume is associated with cognitive decline, and explore factors associated with CP volume in older adults. METHODS We measured CP volume, brain parenchyma, and cerebrospinal fluid (CSF) spaces associated with disproportionately enlarged subarachnoid space hydrocephalus (DESH), an imaging feature of normal-pressure hydrocephalus, in community-dwelling older adults aged ≥ 65 years without dementia. RESULTS In 1,370 participants, lower Mini-Mental State Examination (MMSE) scores were significantly associated with higher CP volume, even after adjusting for DESH-related CSF space and brain parenchymal volume. CP volume was more strongly associated with MMSE scores than DESH-related CSF space and brain parenchymal volume. History of smoking, white matter hyperintensity, enlarged perivascular spaces, age, body mass index, and diabetes mellitus were also associated with increased CP volume. CONCLUSIONS CP volume may be a highly sensitive imaging marker of cognitive decline in community-dwelling older adults without dementia, as it is linked to cognitive decline independently of brain parenchyma and CSF volumes. Our findings emphasize the importance of investigating CP volume increase to maintain cognitive function in older individuals. Accordingly, further longitudinal studies are required.
Collapse
Affiliation(s)
- Yosuke Hidaka
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Mamoru Hashimoto
- Department of Neuropsychiatry, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Takashi Suehiro
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryuji Fukuhara
- Department of Psychiatry, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tomohisa Ishikawa
- Department of Psychiatry, Arao Kokoronosato Hospital, Arao, Japan
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoko Tsunoda
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Geriatric Psychiatry, Mitsugumachi Clinic, Kumamoto, Japan
| | - Asuka Koyama
- Faculty of Social Welfare, Kumamoto Gakuen University, Kumamoto, Japan
| | - Kazuki Honda
- Department of Geriatric Psychiatry, Mitsugumachi Clinic, Kumamoto, Japan
| | - Yusuke Miyagawa
- Department of Psychiatry, Kumamoto Seimei Hospital, Kumamoto, Japan
| | - Kazuhiro Yoshiura
- Department of Frailty Research, Center for Gerontology and Social Science, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Seiji Yuuki
- Department of Psychiatry, Mashiki Hospital, Kumamoto, Japan
| | - Naoto Kajitani
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shuken Boku
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazunari Ishii
- Department of Radiology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Minoru Takebayashi
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
2
|
Manelis A, Hu H, Satz S. The Relationship Between Reduced Hand Dexterity and Brain Structure Abnormality in Older Adults. Geriatrics (Basel) 2024; 9:165. [PMID: 39727824 DOI: 10.3390/geriatrics9060165] [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: 11/12/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Hand dexterity is affected by normal aging and neuroinflammatory processes in the brain. Understanding the relationship between hand dexterity and brain structure in neurotypical older adults may be informative about prodromal pathological processes, thus providing an opportunity for earlier diagnosis and intervention to improve functional outcomes. METHODS this study investigates the associations between hand dexterity and brain measures in neurotypical older adults (≥65 years) using the Nine-Hole Peg Test (9HPT) and magnetic resonance imaging (MRI). RESULTS Elastic net regularized regression revealed that reduced hand dexterity in dominant and non-dominant hands was associated with an enlarged volume of the left choroid plexus, the region implicated in neuroinflammatory and altered myelination processes, and reduced myelin content in the left frontal operculum, the region implicated in motor imagery, action production, and higher-order motor functions. Distinct neural mechanisms underlying hand dexterity in dominant and non-dominant hands included the differences in caudate and thalamic volumes as well as altered cortical myelin patterns in frontal, temporal, parietal, and occipital regions supporting sensorimotor and visual processing and integration, attentional control, and eye movements. Although elastic net identified more predictive features for the dominant vs. non-dominant hand, the feature stability was higher for the latter, thus indicating higher generalizability for the non-dominant hand model. CONCLUSIONS Our findings suggest that the 9HPT for hand dexterity might be a cost-effective screening tool for early detection of neuroinflammatory and neurodegenerative processes. Longitudinal studies are needed to validate our findings in a larger sample and explore the potential of hand dexterity as an early clinical marker.
Collapse
Affiliation(s)
- Anna Manelis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hang Hu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Skye Satz
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
3
|
Bergsland N, Dwyer MG, Zivadinov R. Imaging the Choroid Plexus. Mult Scler 2024; 30:24-29. [PMID: 39658900 DOI: 10.1177/13524585241292965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
The choroid plexus (CP), a highly vascularized structure within the ventricles of the brain, has come under increased scrutiny over the last several years as potentially having a role in the pathophysiology of multiple sclerosis (MS). Originally consider as being only responsible for the production of cerebrospinal fluid, it is now widely recognized that the CP is also involved in immunosurveillance and immune cell trafficking. Histopathology studies have found several immunological changes in donor tissue, including the accumulation of inflammatory cells. These findings have been corroborated by animal studies combining immunohistopathology and magnetic resonance imaging (MRI), showing dynamic changes in CP volume that track immune cell infiltration into the CP itself. Subsequent in vivo studies in persons with MS using MRI have suggested that while CP volume increases very early in the disease, CP inflammation continues to have a role throughout later stages as well. Together with recent advances in image processing methods, the analysis of prospective studies as well as existing datasets will help shed further light on the underlying pathophysiological changes within the CP. Such studies are needed to better understand if the CP may represent a novel therapeutic target to ultimately impact the evolution of the disease.
Collapse
Affiliation(s)
- Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA Center for Biomedical Imaging at the Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
4
|
Stölting A, Vanden Bulcke C, Borrelli S, Bugli C, Du Pasquier R, van Pesch V, Maggi P. Clinical relevance of paramagnetic rim lesion heterogeneity in multiple sclerosis. Ann Clin Transl Neurol 2024; 11:3137-3151. [PMID: 39382072 DOI: 10.1002/acn3.52220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/24/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024] Open
Abstract
OBJECTIVE Previous studies reveal heterogeneity in terms of paramagnetic rim lesions (PRL) associated tissue damage. We investigated the physiopathology and clinical implications of this heterogeneity. METHODS In 103 MS patients (72 relapsing and 31 progressive), brain lesions were manually segmented on 3T 3D-FLAIR and rim visibility was assessed with a visual confidence level score (VCLS) on 3D-EPI phase. Using T1 relaxation time maps, lesions were categorized in long-T1 and short-T1. Lesion age was calculated from time of first gadolinium enhancement (N = 84 lesions). Results on clinical scores were validated in an extended cohort of 167 patients using normalized T1w-MPRAGE lesion values. RESULTS Rim visibility (VCLS analysis) was associated with increasing lesional T1 (P/PFDR < 0.001). Of 1680 analyzed lesions, 427 were categorized as PRL. Long-T1 PRL were older than short-T1 PRL (average 0.8 vs. 2.0 years, P/PFDR = 0.005/0.008), and featured larger lesional volume (P/PFDR < 0.0001) and multi-shell diffusion-measured axonal damage (P/PFDR < 0.0001). The total volume of long-T1-PRL versus PRL showed 2× predictive power for both higher MS disability (EDSS; P/PFDR = 0.003/0.005 vs. P/PFDR = 0.042/0.057) and severity (MSSS; P/PFDR = 0.0006/0.001 vs. P/PFDR = 0.004/0.007). In random forest, having ≥1 long-T1-PRL versus ≥4 PRL showed 2-4× higher performance to predict a higher EDSS and MSSS. In the validation cohort, long-T1 PRL outperformed (~2×) PRL in predicting both EDSS and MSSS. INTERPRETATION PRL show substantial heterogeneity in terms of intralesional tissue damage. More destructive, likely older, long-T1 PRL improve the association with MS clinical scales. This PRL heterogeneity characterization was replicated using standard T1w MRI, highlighting its potential for clinical translation.
Collapse
Affiliation(s)
- Anna Stölting
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium
| | - Colin Vanden Bulcke
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium
- ICTEAM Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Serena Borrelli
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium
- Department of Neurology, Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Brussels, Brussels, Belgium
| | - Céline Bugli
- Plateforme technologique de Support en Méthodologie et Calcul Statistique, Université catholique de Louvain, Brussels, Belgium
| | - Renaud Du Pasquier
- Neurology Service, Department of Clinical Neurosciences, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Vincent van Pesch
- Department of Neurology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Pietro Maggi
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium
- Department of Neurology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
5
|
Hochstetler A, Lehtinen MK. Choroid Plexus as a Mediator of CNS Inflammation in Multiple Sclerosis. Mult Scler 2024; 30:19-23. [PMID: 39503321 PMCID: PMC11634642 DOI: 10.1177/13524585241292974] [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: 12/12/2024]
Abstract
The pathophysiology of multiple sclerosis (MS) remains poorly understood despite decades of tremendous research efforts. Advances in neuroradiography coupled with availability of unbiased approaches including spatial transcriptomics, proteomics, metabolomics, and lipidomics that are compatible with brain and fluid specimens from patients raise hope that discovery of novel disease drivers is on the horizon. Once thought to be little more than salty bathwater, our modern understanding of cerebrospinal fluid (CSF) suggests the CSF as a compelling, critical regulator of brain function in health and disease. Recent studies in the field have reinvigorated interest in CSF as a medium to better understand MS and to deliver disease-modifying therapies. In turn, the choroid plexus, an epithelial tissue located within each brain ventricle that regulates CSF and forms a key blood-CSF barrier, is uniquely positioned to orchestrate neuroinflammation associated with MS. In this perspective review, we will discuss what is known about the choroid plexus as a conductor of immune responses and how it may propagate neuroinflammation associated with MS via the CSF.
Collapse
Affiliation(s)
- Alexandra Hochstetler
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Bravi B, Paolini M, Maccario M, Milano C, Raffaelli L, Melloni EMT, Zanardi R, Colombo C, Benedetti F. Abnormal choroid plexus, hippocampus, and lateral ventricles volumes as markers of treatment-resistant major depressive disorder. Psychiatry Clin Neurosci 2024. [PMID: 39563010 DOI: 10.1111/pcn.13764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/09/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024]
Abstract
AIM One-third of patients with major depressive disorder (MDD) do not achieve full remission and have high relapse rates even after treatment, leading to increased medical costs and reduced quality of life and health status. The possible specificity of treatment-resistant depression (TRD) neurobiology is still under investigation, with risk factors such as higher inflammatory markers being identified. Given recent findings on the role of choroid plexus (ChP) in neuroinflammation and hippocampus in treatment response, the aim of the present study was to evaluate inflammatory- and trophic-related differences in these regions along with ventricular volumes among patients with treatment-sensitive depression (TSD), TRD, and healthy controls (HCs). METHODS ChP, hippocampal, and ventricular volumes were assessed in 197 patients with MDD and 58 age- and sex-matched HCs. Volumes were estimated using FreeSurfer 7.2. Treatment resistance status was defined as failure to respond to at least two separate antidepressant treatments. Region of interest volumes were then compared among groups. RESULTS We found higher ChP volumes in patients with TRD compared with patients with TSD and HCs. Our results also showed lower hippocampal volumes and higher lateral ventricular volumes in TRD compared with both patients without TRD and HCs. CONCLUSIONS These findings corroborate the link between TRD and neuroinflammation, as ChP volume could be considered a putative marker of central immune activity. The lack of significant differences in all of the region of interest volumes between patients with TSD and HCs may highlight the specificity of these features to TRD, possibly providing new insights into the specific neurobiological underpinnings of this condition.
Collapse
Affiliation(s)
- Beatrice Bravi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Marco Paolini
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Melania Maccario
- University Vita-Salute San Raffaele, Milan, Italy
- Mood Disorders Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Chiara Milano
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Laura Raffaelli
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Elisa Maria Teresa Melloni
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Raffaella Zanardi
- University Vita-Salute San Raffaele, Milan, Italy
- Mood Disorders Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Cristina Colombo
- University Vita-Salute San Raffaele, Milan, Italy
- Mood Disorders Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
7
|
Grasso EA, Bloy L, Kaplan P, Bar-Or A, Yeh EA, Arnold DL, Narayanan S, Marrie RA, Fadda G, Banwell BL. Choroid Plexus Volume in Pediatric-Onset Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200319. [PMID: 39442038 PMCID: PMC11502105 DOI: 10.1212/nxi.0000000000200319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/16/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND OBJECTIVES Recent studies suggest that the choroid plexus (CP) may function as a site of access of inflammatory cells into the CNS in multiple sclerosis (MS). Pediatric-onset MS (POMS) is characterized by a high inflammatory burden, as evidenced by a high relapse rate and volume of T2 lesions, making patients with POMS an informative population to evaluate choroid plexus volume (CPV). The objectives of the study were (1) to evaluate CPV at symptom onset in participants with POMS compared with healthy controls (HCs); (2) to evaluate changes in CPV in the first year of disease in participants with POMS; and (3) to evaluate associations between CPV, brain volumes, relapse activity, and disability in participants with POMS. METHODS Baseline 1.5T MRI scans were acquired from 23 participants with POMS and 23 age-matched and sex-matched HCs; 18 participants with POMS also had 12-month follow-up MRI scans. The CP of the lateral ventricles was segmented manually. CP and brain structure volumes were normalized for total intracranial volume. The number of relapses, T2 and gadolinium-enhancing T1 lesion counts, and Expanded Disability Status Scale (EDSS) scores at 12 months were also analyzed. Baseline CPVs were compared between groups using the Wilcoxon exact test, and CPV change from baseline to 12 months in participants with POMS was compared using the Wilcoxon signed-rank test. The relationship between CPV and brain volumetric measures, T2 lesion volumes, lesion count, number of relapses, and EDSS scores was assessed through Spearman correlation. RESULTS The median normalized CPV was 1.51 × 10-3 (interquartile range [IQR]: 1.32-1.76) in POMS baseline scans and 1.21 × 10-3 (IQR: 1.1-1.47) in HC scans (p = 0.001). CPV did not significantly change at 12 months in the 18 participants with POMS with follow-up scans (p = 0.352). CPV in participants with POMS and HCs correlated with lateral ventricular volume (p = 0.012 for both groups) but did not correlate with brain and T2 lesion volumes or lesion count at baseline, nor with relapse activity or EDSS scores at 12 months in participants with POMS. DISCUSSION CPV measured at baseline is greater in participants with POMS than in HCs. Baseline CPV did not predict higher disease activity or worse neurologic outcomes over 1 year. While higher CPV may be an early feature of inflammation in MS, its strong correlation with ventricular volumes could also reflect enlargement secondary to the mechanical attachment of CP to the ventricular wall.
Collapse
Affiliation(s)
- Eleonora A Grasso
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Luke Bloy
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Phillip Kaplan
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Amit Bar-Or
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - E Ann Yeh
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Douglas L Arnold
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Sridar Narayanan
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Ruth Ann Marrie
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Giulia Fadda
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Brenda L Banwell
- From the Departments of Neurology (E.A.G., P.K., B.L.B.), Radiology (L.B.), Children's Hospital of Philadelphia, PA; Department of Neurology (A.B.-O.), University of Pennsylvania, PA; Division of Neurology (E.A.Y.), The Hospital for Sick Children, Toronto, Canada; McConnell Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, Montreal, Canada; Department of Neurology (S.N.), McGill University, Montreal, Canada; Departments of Medicine and Community Health Sciences (R.A.M.), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; and Department of Medicine (G.F.), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| |
Collapse
|
8
|
Kolahi S, Zarei D, Issaiy M, Shakiba M, Azizi N, Firouznia K. Choroid plexus volume changes in multiple sclerosis: insights from a systematic review and meta-analysis of magnetic resonance imaging studies. Neuroradiology 2024; 66:1869-1886. [PMID: 39105769 DOI: 10.1007/s00234-024-03439-3] [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: 04/29/2024] [Accepted: 07/27/2024] [Indexed: 08/07/2024]
Abstract
PURPOSE Multiple sclerosis (MS) is a chronic autoimmune disease characterized by the destruction of the myelin sheath within the central nervous system. The etiology of MS involves a complex interplay of genetic, environmental, and immunological factors. Recent studies indicated the potential role of the choroid plexus (CP) in the pathogenesis and progression of MS. This systematic review aims to assess existing research on the volume alterations of the CP in MS patients compared to the normal population. METHODS A comprehensive search was conducted across databases including PubMed, Embase, Scopus, and Web of Science up to June 2024. Data from the included studies were synthesized using a meta-analytical approach with a random-effects model, assessing heterogeneity with the I2 and Tau-squared indices. RESULTS We included 17 studies in this systematic review. The meta-analysis, which included data from eight studies reporting CP volume relative to TIV, found a statistically significant increase in CP volume in MS patients compared to healthy controls (HCs). The SMD was 0.77 (95% CI: 0.61 to 0.93), indicating a large effect size. This analysis showed no heterogeneity (I² = 0%). A separate meta-analysis was conducted using five studies that reported CP volume as normalized volume, resulting in an SMD of 0.63 (95% CI: 0.2-1.06). CONCLUSION This study demonstrates an increase in CP volume among MS patients compared to HCs, implying the potential involvement of CP in MS pathogenesis and/or progression. These results show that CP might serve as a radiological indicator in the diagnosis and prognosis of MS.
Collapse
Affiliation(s)
- Shahriar Kolahi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Diana Zarei
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Mahbod Issaiy
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Madjid Shakiba
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Narges Azizi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Kavous Firouznia
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Visani V, Veronese M, Pizzini FB, Colombi A, Natale V, Marjin C, Tamanti A, Schubert JJ, Althubaity N, Bedmar-Gómez I, Harrison NA, Bullmore ET, Turkheimer FE, Calabrese M, Castellaro M. ASCHOPLEX: A generalizable approach for the automatic segmentation of choroid plexus. Comput Biol Med 2024; 182:109164. [PMID: 39326265 DOI: 10.1016/j.compbiomed.2024.109164] [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: 04/09/2024] [Revised: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND The Choroid Plexus (ChP) plays a vital role in brain homeostasis, serving as part of the Blood-Cerebrospinal Fluid Barrier, contributing to brain clearance pathways and being the main source of cerebrospinal fluid. Since the involvement of ChP in neurological and psychiatric disorders is not entirely established and currently under investigation, accurate and reproducible segmentation of this brain structure on large cohorts remains challenging. This paper presents ASCHOPLEX, a deep-learning tool for the automated segmentation of human ChP from structural MRI data that integrates existing software architectures like 3D UNet, UNETR, and DynUNet to deliver accurate ChP volume estimates. METHODS Here we trained ASCHOPLEX on 128 T1-w MRI images comprising both controls and patients with Multiple Sclerosis. ASCHOPLEX's performances were evaluated using traditional segmentation metrics; manual segmentation by experts served as ground truth. To overcome the generalizability problem that affects data-driven approaches, an additional fine-tuning procedure (ASCHOPLEXtune) was implemented on 77 T1-w PET/MRI images of both controls and depressed patients. RESULTS ASCHOPLEX showed superior performance compared to commonly used methods like FreeSurfer and Gaussian Mixture Model both in terms of Dice Coefficient (ASCHOPLEX 0.80, ASCHOPLEXtune 0.78) and estimated ChP volume error (ASCHOPLEX 9.22%, ASCHOPLEXtune 9.23%). CONCLUSION These results highlight the high accuracy, reliability, and reproducibility of ASCHOPLEX ChP segmentations.
Collapse
Affiliation(s)
- Valentina Visani
- Department of Information Engineering, University of Padova, Padova, Italy.
| | - Mattia Veronese
- Department of Information Engineering, University of Padova, Padova, Italy; Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Francesca B Pizzini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy.
| | | | - Valerio Natale
- Department of Diagnostic and Public Health, University of Verona, Verona, Italy.
| | - Corina Marjin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| | - Agnese Tamanti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| | - Julia J Schubert
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Noha Althubaity
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Radiological Sciences, College of Applied Medical Science, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia.
| | - Inés Bedmar-Gómez
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Neil A Harrison
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Cardiff, UK.
| | - Edward T Bullmore
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK; Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK; Immuno-Psychiatry, Immuno-Inflammation Therapeutic Area Unit, GlaxoSmithKline R&D, Stevenage, UK.
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Massimiliano Calabrese
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| | - Marco Castellaro
- Department of Information Engineering, University of Padova, Padova, Italy.
| |
Collapse
|
10
|
Hashemi E, Srivastava IN, Aguirre A, Yoseph ET, Kaushal E, Awani A, Ryu JK, Akassoglou K, Talebian S, Chu P, Pisani L, Musolino P, Steinman L, Doyle K, Robinson WH, Sharpe O, Cayrol R, Orchard PJ, Lund T, Vogel H, Lenail M, Han MH, Bonkowsky JL, Van Haren KP. A Novel Mouse Model for Cerebral Inflammatory Demyelination in X-Linked Adrenoleukodystrophy: Insights into Pathogenesis and Potential Therapeutic Targets. Ann Neurol 2024. [PMID: 39467011 DOI: 10.1002/ana.27117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVE X-linked adrenoleukodystrophy (ALD) is caused by mutations in ABCD1, a peroxisomal gene. More than half of males with an ABCD1 mutation develop inflammatory cerebral demyelination (cALD), but underlying mechanisms remain unknown and therapies are limited. We sought to develop and characterize a mouse model of cALD to facilitate study of disease mechanisms and therapy development. METHODS We used immunoassays and immunohistochemistry to assess novel (interleukin 18 [IL-18]) and established molecular markers in cerebrospinal fluid (CSF) and postmortem brain tissue from cALD patients. We generated a cALD phenotype in Abcd1-knockout mice using a 2-hit method that combines cuprizone and experimental autoimmune encephalomyelitis models. We then used magnetic resonance imaging (MRI) and immunohistochemistry to assess the fidelity of cALD molecular markers in the mice. RESULTS Human and mouse cALD lesions shared histologic features of myelin phagocytosis, myelin loss, abundant microglial activation, T and B-cell infiltration, and astrogliosis. Compared to wild-type controls, Abcd1-knockout mice displayed more cerebral demyelination, blood-brain barrier disruption, and perivascular immune cell infiltration. This enhanced inflammatory response was associated with higher levels of fibrin deposition, oxidative stress, demyelination, and axonal injury. IL-18 immunoreactivity co-localized with perivascular monocytes/macrophages in both human and mouse brain tissue. In cALD patients, CSF IL-18 levels correlated with MRI lesion severity. INTERPRETATION Our results suggest loss of Abcd1 function in mice predisposes to more severe blood-brain barrier disruption, cerebral inflammation driven by the infiltration of peripheral immune cells, demyelination, and axonal damage, replicating human cALD features. This novel mouse model could shed light on cALD mechanisms and accelerate cALD therapy development. ANN NEUROL 2024.
Collapse
Affiliation(s)
- Ezzat Hashemi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Isha N Srivastava
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Alejandro Aguirre
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Ezra T Yoseph
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Esha Kaushal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Avni Awani
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Jae K Ryu
- Gladstone Institute for Neurological Disease, San Francisco, CA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Katerina Akassoglou
- Gladstone Institute for Neurological Disease, San Francisco, CA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Shahrzad Talebian
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Pauline Chu
- Stanford Human Research Histology Core, Stanford University School of Medicine, Stanford, CA
| | - Laura Pisani
- Department of Radiology, Stanford University School of Medicine Stanford, Stanford, CA
| | - Patricia Musolino
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Kristian Doyle
- Department of Immunobiology, University of Arizona, Tucson, AZ
| | - William H Robinson
- Department of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA
| | - Orr Sharpe
- Department of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA
| | - Romain Cayrol
- Department of Pathology, Clinical Department of Laboratory Medicine, University of Montreal, Quebec, Canada
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN
| | - Troy Lund
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN
| | - Hannes Vogel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Max Lenail
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - May H Han
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Joshua L Bonkowsky
- Division of Pediatric Neurology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT
- Brain and Spine Center, Primary Children's Hospital, Salt Lake City, UT
- Primary Children's Center for Personalized Medicine, Salt Lake City, UT
| | - Keith P Van Haren
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
11
|
Sun S, Chen Y, Yun Y, Zhao B, Ren Q, Sun X, Meng X, Yan C, Lin P, Liu S. Elevated peripheral inflammation is associated with choroid plexus enlargement in independent sporadic amyotrophic lateral sclerosis cohorts. Fluids Barriers CNS 2024; 21:83. [PMID: 39434103 PMCID: PMC11492712 DOI: 10.1186/s12987-024-00586-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Using neuroimaging techniques, growing evidence has suggested that the choroid plexus (CP) volume is enlarged in multiple neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Notably, the CP has been suggested to play an important role in inflammation-induced CNS damage under disease conditions. However, to our knowledge, no study has investigated the relationships between peripheral inflammation and CP volume in sporadic ALS patients. Thus, in this study, we aimed to verify CP enlargement and explore its association with peripheral inflammation in vivo in independent ALS cohorts. METHODS Based on structural MRI data, CP volume was measured using Gaussian mixture models and further manually corrected in two independent cohorts of sporadic ALS patients and healthy controls (HCs). Serum inflammatory protein levels were measured using a novel high-sensitivity Olink proximity extension assay (PEA) technique. Xtreme gradient boosting (XGBoost) was used to explore the contribution of peripheral inflammatory factors to CP enlargement. Then, partial correlation analyses were performed. RESULTS CP volumes were significantly higher in ALS patients than in HCs in the independent cohorts. Compared with HCs, serum levels of CRP, IL-6, CXCL10, and 35 other inflammatory factors were significantly increased in ALS patients. Using the XGBoost approach, we established a model-based importance of features, and the top three predictors of CP volume in ALS patients were CRP, IL-6, and CXCL10 (with gains of 0.24, 0.18, and 0.15, respectively). Correlation analyses revealed that CRP, IL-6, and CXCL10 were significantly associated with CP volume in ALS patients (r = 0.462 ∼ 0.636, p < 0.001). CONCLUSION Our study is the first to reveal a consistent and replicable contribution of peripheral inflammation to CP enlargement in vivo in sporadic ALS patients. Given that CP enlargement has been recently detected in other brain diseases, these findings should consider extending to other disease conditions with a peripheral inflammatory component.
Collapse
Affiliation(s)
- Sujuan Sun
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China
| | - Yujing Chen
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China
| | - Yan Yun
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Bing Zhao
- Department of Neurology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, China
| | - Qingguo Ren
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, China
| | - Xiaohan Sun
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China
| | - Xiangshui Meng
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, China
| | - Chuanzhu Yan
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Pengfei Lin
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China.
| | - Shuangwu Liu
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China.
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China.
| |
Collapse
|
12
|
Jakimovski D, Zivadinov R, Ramanathan M, Weinstock-Guttman B, Tavazzi E, Dwyer MG, Bergsland N. Greater humoral EBV response may be associated with choroid plexus inflammation in progressive MS. J Neurovirol 2024:10.1007/s13365-024-01231-w. [PMID: 39420132 DOI: 10.1007/s13365-024-01231-w] [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: 06/16/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
Choroid plexus (CP) inflammation can be quantified in vivo with MRI in people with multiple sclerosis (pwMS). It remains unknown whether Epstein Barr Virus (EBV) is related to CP changes. Total of 170 pwMS (116 relapsing-remitting; RRMS and 54 progressive MS; PMS) underwent MRI examination and measurement of humoral anti-EBV response. CP volume and CP pseudo-T2 (pT2), a relaxation time indicative of edema and neuroinflammation, were measured. Moreover, anti-EBV nuclear antigen-1 (EBNA-1) IgG and anti-EBV capsid antigen (VCA) IgG antibodies were measured. The PMS group had greater CP pT2 value when compared to RRMS (1120ms vs. 954ms, p = 0.037). After adjusting for age and therapy effects, higher CP pT2 values were associated with higher anti-EBNA-1 IgG levels only in PMS (r = 0.352, p = 0.015). Higher Anti-EBV humoral response in pwMS may be associated with increased CP neuroinflammatory changes and may be more relevant for the later chronic stage of the disease. Large-scale studies should investigate whether these findings are generalizable to all types of progressive MS.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 100 High St, Buffalo, NY, 14203, USA.
- Wynn Hospital, Mohawk Valley Health System, Utica, NY, 13502, USA.
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 100 High St, Buffalo, NY, 14203, USA
- Center for Biomedical Imaging at the Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Eleonora Tavazzi
- Multiple Sclerosis Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 100 High St, Buffalo, NY, 14203, USA
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 100 High St, Buffalo, NY, 14203, USA
| |
Collapse
|
13
|
Rubin M, Preziosa P, Margoni M, Meani A, Pagani E, Corazzolla G, Storelli L, Mistri D, Filippi M, Rocca MA. Dynamics of choroid plexus volume is associated with the presence and development of fatigue in multiple sclerosis. J Neurol Neurosurg Psychiatry 2024:jnnp-2024-334913. [PMID: 39389772 DOI: 10.1136/jnnp-2024-334913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Immune-mediated processes are implicated in the pathogenesis of fatigue, a common symptom in multiple sclerosis (MS). The choroid plexus (CP) regulates central nervous system (CNS) immune homeostasis and undergoes volumetric modifications possibly contributing to MS-related fatigue. We explored the association between MS-related CP volume changes and fatigue dynamics. METHOD Eighty-five patients with MS and 68 healthy controls (HC) underwent brain 3T MRI, neurological evaluation and Modified Fatigue Impact Scale (MFIS) at two timepoints (median follow-up=1.4 years). Normalised brain and regional grey matter (GM) volumes were obtained using FSL-SIENAx, FIRST, SIENA and tensor-based morphometry. CP volumes were quantified with in-house methods, and longitudinal changes were analysed using linear mixed models. RESULTS At baseline, 25 (29%) patients with MS had fatigue (f-MS) (MFIS ≥38). Compared with HC, patients with MS had significantly higher brain T2-lesion volume, lower brain, deep GM, cortical volumes and higher CP volume (false discovery rate (FDR)-p ≤0.024). Compared with non-fatigued (nf-MS) patients, f-MS were older, more disabled (FDR-p ≤0.002) and showed numerically higher CP volume (FDR-p=0.076). At follow-up, 41 (68%) nf-MS remained non-fatigued (nf-FU-MS) and 19 (32%) developed fatigue (f-FU-MS). Patients with MS showed higher brain and deep GM atrophy rates versus HC (FDR-p ≤0.048), whereas clinical, lesional and brain volumetric changes were not significantly different among MS groups (FDR-p ≥0.287). CP volume significantly increased in all MS groups compared with HC (FDR-p ≤0.043), with greater enlargement in f-FU-MS versus nf-FU-MS (FDR-p=0.048). CONCLUSIONS Larger CP and greater enlargement are associated with the presence and development of fatigue in MS, likely reflecting dynamic inflammatory states within the CNS, supporting the immunological contribution to MS-related fatigue.
Collapse
Affiliation(s)
- Martina Rubin
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Neurology Unit, IRCCS Ospedale San Raffaele, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Neurology Unit, IRCCS Ospedale San Raffaele, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Monica Margoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Neurology Unit, IRCCS Ospedale San Raffaele, Milano, Italy
- Neurorehabilitation Unit, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Alessandro Meani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Elisabetta Pagani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Gianluca Corazzolla
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Loredana Storelli
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Damiano Mistri
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Neurology Unit, IRCCS Ospedale San Raffaele, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
- Neurorehabilitation Unit, IRCCS Ospedale San Raffaele, Milano, Italy
- Neurophysiology Service, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Neurology Unit, IRCCS Ospedale San Raffaele, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
14
|
Bouhrara M, Walker KA, Alisch JSR, Gong Z, Mazucanti CH, Lewis A, Moghekar AR, Turek L, Collingham V, Shehadeh N, Fantoni G, Kaileh M, Bergeron CM, Bergeron J, Resnick SM, Egan JM. Association of Plasma Markers of Alzheimer's Disease, Neurodegeneration, and Neuroinflammation with the Choroid Plexus Integrity in Aging. Aging Dis 2024; 15:2230-2240. [PMID: 38300640 PMCID: PMC11346414 DOI: 10.14336/ad.2023.1226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024] Open
Abstract
The choroid plexus (CP) is a vital brain structure essential for cerebrospinal fluid (CSF) production. Moreover, alterations in the CP's structure and function are implicated in molecular conditions and neuropathologies including multiple sclerosis, Alzheimer's disease, and stroke. Our goal is to provide the first characterization of the association between variation in the CP microstructure and macrostructure/volume using advanced magnetic resonance imaging (MRI) methodology, and blood-based biomarkers of Alzheimer's disease (Aß42/40 ratio; pTau181), neuroinflammation and neuronal injury (GFAP; NfL). We hypothesized that plasma biomarkers of brain pathology are associated with disordered CP structure. Moreover, since cerebral microstructural changes can precede macrostructural changes, we also conjecture that these differences would be evident in the CP microstructural integrity. Our cross-sectional study was conducted on a cohort of 108 well-characterized individuals, spanning 22-94 years of age, after excluding participants with cognitive impairments and non-exploitable MR imaging data. Established automated segmentation methods were used to identify the CP volume/macrostructure using structural MR images, while the microstructural integrity of the CP was assessed using our advanced quantitative high-resolution MR imaging of longitudinal and transverse relaxation times (T1 and T2). After adjusting for relevant covariates, positive associations were observed between pTau181, NfL and GFAP and all MRI metrics. These associations reached significance (p<0.05) except for CP volume vs. pTau181 (p=0.14), CP volume vs. NfL (p=0.35), and T2 vs. NFL (p=0.07). Further, negative associations between Aß42/40 and all MRI metrics were observed but reached significance only for Aß42/40 vs. T2 (p=0.04). These novel findings demonstrate that reduced CP macrostructural and microstructural integrity is positively associated with blood-based biomarkers of AD pathology, neurodegeneration/neuroinflammation and neurodegeneration. Degradation of the CP structure may co-occur with AD pathology and neuroinflammation ahead of clinically detectable cognitive impairment, making the CP a potential structure of interest for early disease detection or treatment monitoring.
Collapse
Affiliation(s)
- Mustapha Bouhrara
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Joseph S. R. Alisch
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Zhaoyuan Gong
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Caio H. Mazucanti
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Alexandria Lewis
- Johns Hopkins University School of Medicine, Baltimore, 21224 MD, USA.
| | - Abhay R. Moghekar
- Johns Hopkins University School of Medicine, Baltimore, 21224 MD, USA.
| | - Lisa Turek
- Clinical Research Core, Baltimore, MD 21224, USA.
| | | | | | | | - Mary Kaileh
- Clinical Research Core, Baltimore, MD 21224, USA.
| | - Christopher M. Bergeron
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Jan Bergeron
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Josephine M. Egan
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
15
|
Rau A, Gonzalez-Escamilla G, Schroeter N, Othman A, Dressing A, Weiller C, Urbach H, Reisert M, Groppa S, Hosp JA. Inflammation-Triggered Enlargement of Choroid Plexus in Subacute COVID-19 Patients with Neurological Symptoms. Ann Neurol 2024; 96:715-725. [PMID: 38934493 DOI: 10.1002/ana.27016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE To investigate whether choroid plexus volumes in subacute coronavirus disease 2019 (COVID-19) patients with neurological symptoms could indicate inflammatory activation or barrier dysfunction and assess their association with clinical data. METHODS Choroid plexus volumes were measured in 28 subacute COVID-19 patients via cerebral magnetic resonance imaging (MRI), compared with those in infection-triggered non-COVID-19 encephalopathy patients (n = 25), asymptomatic individuals after COVID-19 (n = 21), and healthy controls (n = 21). Associations with inflammatory serum markers (peak counts of leukocytes, C-reactive protein [CRP], interleukin 6), an MRI-based marker of barrier dysfunction (CSF volume fraction [V-CSF]), and clinical parameters like olfactory performance and cognitive scores (Montreal Cognitive Assessment) were investigated. RESULTS COVID-19 patients showed significantly larger choroid plexus volumes than control groups (p < 0.001, η2 = 0.172). These volumes correlated significantly with peak leukocyte levels (p = 0.001, Pearson's r = 0.621) and V-CSF (p = 0.009, Spearman's rho = 0.534), but neither with CRP nor interleukin 6. No significant correlations were found with clinical parameters. INTERPRETATION In patients with subacute COVID-19, choroid plexus volume is a marker of central nervous system inflammation and barrier dysfunction in the presence of neurologic symptoms. The absence of plexus enlargement in infection-triggered non-COVID-19 encephalopathy suggests a specific severe acute respiratory syndrome coronavirus 2 effect. This study also documents an increase in choroid plexus volume for the first time as a parainfectious event. ANN NEUROL 2024;96:715-725.
Collapse
Affiliation(s)
- Alexander Rau
- Department of Neuroradiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gabriel Gonzalez-Escamilla
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nils Schroeter
- Department of Neurology and Clinical Neuroscience, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ahmed Othman
- Department of Neuroradiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Andrea Dressing
- Department of Neurology and Clinical Neuroscience, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius Weiller
- Department of Neurology and Clinical Neuroscience, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Horst Urbach
- Department of Neuroradiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Reisert
- Department of Medical Physics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sergiu Groppa
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jonas A Hosp
- Department of Neurology and Clinical Neuroscience, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
16
|
Landes-Château C, Ricigliano VA, Mondot L, Thouvenot E, Labauge P, Louapre C, Zéphir H, Durand-Dubief F, Le Page E, Siva A, Cohen M, Yazdan Panah A, Azevedo CJ, Okuda DT, Stankoff B, Lebrun-Frénay C. Choroid plexus enlargement correlates with periventricular pathology but not with disease activity in radiologically isolated syndrome. Mult Scler 2024; 30:1278-1289. [PMID: 39246289 DOI: 10.1177/13524585241272943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
BACKGROUND Choroid plexus (ChP) enlargement is an emerging radiological biomarker in multiple sclerosis (MS). OBJECTIVES This study aims to assess ChP volume in a large cohort of patients with radiologically isolated syndrome (RIS) versus healthy controls (HC) and explore its relationship with other brain volumes, disease activity, and biological markers. METHODS RIS individuals were included retrospectively and compared with HC. ChPs were automatically segmented using an in-house automated algorithm and manually corrected. RESULTS A total of 124 patients fulfilled the 2023 RIS criteria, and 55 HCs were included. We confirmed that ChPs are enlarged in RIS versus HC (mean (±SD) normalized ChP volume: 17.24 (±4.95) and 11.61 (±3.58), respectively, p < 0.001). Larger ChPs were associated with more periventricular lesions (ρ = 0.26; r2 = 0.27; p = 0.005 for the correlation with lesion volume, and ρ = 0.2; r2 = 0.21; p = 0.002 for the correlation with lesion number) and lower thalamic volume (ρ = -0.38; r2 = 0.44; p < 0.001), but not with lesions in other brain regions. Conversely, ChP volume did not correlate with biological markers. No significant difference in ChP volume was observed between subjects who presented or did not have a clinical event or between those with or without imaging disease activity. CONCLUSIONS This study provides evidence that ChP volume is higher in RIS and is associated with measures reflecting periventricular pathology but does not correlate with biological, radiological, or clinical markers of disease activity.
Collapse
Affiliation(s)
| | - Vito Ag Ricigliano
- Paris Brain Institute-ICM, CNRS, Inserm, Neurology Department, Pitié-Salpêtrière Hospital, Sorbonne Université, AP-HP, Paris, France
| | | | - Eric Thouvenot
- IGF, University Montpellier, CNRS, INSERM, Montpellier, France
| | - Pierre Labauge
- Centre hospitalier universitaire de Montpellier, Montpellier, France
| | - Céline Louapre
- Paris Brain Institute-ICM, CNRS, Inserm, Neurology Department, Pitié-Salpêtrière Hospital, Sorbonne Université, AP-HP, Paris, France
| | - Hélène Zéphir
- University of Lille, INSERM U 1172, CHU of Lille, Lille, France
| | | | | | - Aksel Siva
- Cerrahpasa School of Medicine, Istanbul University, Istanbul, Turkiye
| | - Mikael Cohen
- Université Côte d'Azur, UMR2CA (URRIS), Nice, France
| | - Arya Yazdan Panah
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, France
| | - Christina J Azevedo
- Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Darin T Okuda
- The University of Texas Southwestern Medical Center, Peter O'Donnell Jr. Brain Institute, Dallas, TX, USA
| | - Bruno Stankoff
- Paris Brain Institute-ICM, CNRS, Inserm, Neurology Department, Pitié-Salpêtrière Hospital, Sorbonne Université, AP-HP, Paris, France
| | | |
Collapse
|
17
|
Agarwal N, Fan A, Huang X, Dehkharghani S, van der Kolk A. ISMRM Clinical Focus Meeting 2023: "Imaging the Fire in the Brain". J Magn Reson Imaging 2024. [PMID: 39193867 DOI: 10.1002/jmri.29587] [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: 05/24/2024] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 08/29/2024] Open
Abstract
Set during the Annual Meeting of the International Society for Magnetic Resonance in Medicine (ISMRM), the "Clinical Focus Meeting" (CFM) aims to bridge the gap between innovative magnetic resonance imaging (MRI) scientific research and daily patient care. This initiative is dedicated to maximizing the impact of MRI technology on healthcare outcomes for patients. At the 2023 Annual Meeting, clinicians and scientists from across the globe were invited to discuss neuroinflammation from various angles (entitled "Imaging the Fire in the Brain"). Topics ranged from fundamental mechanisms and biomarkers of neuroinflammation to the role of different contrast mechanisms, including both proton and non-proton techniques, in brain tumors, autoimmune disorders, and pediatric neuroinflammatory diseases. Discussions also delved into how systemic inflammation can trigger neuroinflammation and the role of the gut-brain axis in causing brain inflammation. Neuroinflammation arises from various external and internal factors and serves as a vital mechanism to mitigate tissue damage and provide neuroprotection. Nonetheless, excessive neuroinflammatory responses can lead to significant tissue injury and subsequent neurological impairments. Prolonged neuroinflammation can result in cellular apoptosis and neurodegeneration, posing severe consequences. MRI can be used to visualize these consequences, by detecting blood-brain barrier damage, characterizing brain lesions, quantifying edema, and identifying specific metabolites. It also facilitates monitoring of chronic changes in both the brain and spinal cord over time, potentially leading to better patient outcomes. This paper represents a summary of the 2023 CFM, and is intended to guide the enthusiastic MR user to several key and novel sequences that MRI offers to image pathophysiologic processes underlying acute and chronic neuroinflammation. EVIDENCE LEVEL: 5 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Nivedita Agarwal
- Diagnostic Imaging and Neuroradiology Unit, IRCCS Scientific Institute E. Medea, Bosisio Parini, Lecco, Italy
| | - Audrey Fan
- Department of Neurology, University of California Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Xiaoqi Huang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Seena Dehkharghani
- Department of Radiology, Albert Einstein College of Medicine-Montefiore Health, New York, New York, USA
| | - Anja van der Kolk
- Department of Medical Imaging, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
18
|
Wang X, Liu S, Yan Z, Yin F, Feng J, Liu H, Liu Y, Li Y. Radiomics Nomograms Based on Multi-sequence MRI for Identifying Cognitive Impairment and Predicting Cognitive Progression in Relapsing-Remitting Multiple Sclerosis. Acad Radiol 2024:S1076-6332(24)00591-9. [PMID: 39198138 DOI: 10.1016/j.acra.2024.08.026] [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: 07/08/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
RATIONALE AND OBJECTIVES To build radiomics nomograms based on multi-sequence MRI to facilitate the identification of cognitive impairment (CI) and prediction of cognitive progression (CP) in patients with relapsing-remitting multiple sclerosis (RRMS). MATERIALS AND METHODS We retrospectively included two RRMS cohorts with multi-sequence MRI and Symbol Digit Modalities Test (SDMT) data: dataset1 (n = 149, for training and validation) and dataset2 (n = 29, for external validation). 80 patients of dataset1 had a 2-year follow-up SDMT. CI and CP were evaluated using SDMT scores at baseline and follow-up. The included DIR sequence aided in identifying cortical lesions. Lesion radiomics and structural features were extracted and selected from multi-sequence MRI, followed by the computation of radiomics and structural scores. The nomogram was developed through multivariate logistic regression, integrating clinical data, radiomics, and structural scores to identify CI in patients. Moreover, a similar method was employed to further construct a nomogram predicting CP in patients. RESULTS The nomogram demonstrated superior performance in identifying patients with CI, with area under the curve (AUC) values of 0.937 (95% Conf. Interval: 0.898-0.975) and 0.876 (0.810-0.943) in internal and external validation sets, compared to models solely based on clinical data, lesion radiomics, and structural features. Furthermore, another nomogram constructed in predicting CP also exhibited outstanding performance, with an AUC value of 0.969 (0.875-1.000) in the validation set. CONCLUSION These nomograms, integrating clinical data, multi-sequence lesions radiomics, and structural features, enable more effective identification of CI and early prediction of CP in RRMS patients, providing important support for clinical decision-making.
Collapse
Affiliation(s)
- Xiaohua Wang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Shangqing Liu
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Zichun Yan
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Feiyue Yin
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jinzhou Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hao Liu
- Yizhun Medical AI, Beijing 100000, China
| | - Yanbing Liu
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
19
|
Wang X, Wang X, Yan Z, Yin F, Li Y, Liu X, Liu Y. Enhanced choroid plexus segmentation with 3D UX-Net and its association with disease progression in multiple sclerosis. Mult Scler Relat Disord 2024; 88:105750. [PMID: 38986172 DOI: 10.1016/j.msard.2024.105750] [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: 03/25/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND The choroid plexus (CP) is suggested to be closely associated with the neuroinflammation of multiple sclerosis (MS). Segmentation based on deep learning (DL) could facilitate rapid and reproducible volume assessment of the CP, which is crucial for elucidating its role in MS. PURPOSE To develop a reliable DL model for the automatic segmentation of CP, and further validate its clinical significance in MS. METHODS The 3D UX-Net model (3D U-Net used for comparison) was trained and validated on T1-weighted MRI from a cohort of 216 relapsing-remitting MS (RRMS) patients and 75 healthy subjects. Among these, 53 RRMS with baseline and 2-year follow-up scans formed an internal test set (dataset1b). Another 58 RRMS from multi-center data served as an external test set (dataset2). Dice coefficient was computed to assess segmentation performance. Compare the correlation of CP volume obtained through automatic and manual segmentation with clinical outcomes in MS. Disability and cognitive function of patients were assessed using the Expanded Disability Status Scale (EDSS) and Symbol Digit Modalities Test (SDMT). RESULTS The 3D UX-Net model achieved Dice coefficients of 0.875 ± 0.030 and 0.870 ± 0.044 for CP segmentation on dataset1b and dataset2, respectively, outperforming 3D U-Net's scores of 0.809 ± 0.098 and 0.601 ± 0.226. Furthermore, CP volumes segmented by the 3D UX-Net model aligned consistently with clinical outcomes compared to manual segmentation. In dataset1b, both manual and automatic segmentation revealed a significant positive correlation between normalized CP volume (nCPV) and EDSS scores at baseline (manual: r = 0.285, p = 0.045; automatic: r = 0.287, p = 0.044) and a negative correlation with SDMT scores (manual: r = -0.331, p = 0.020; automatic: r = -0.329, p = 0.021). In dataset2, similar correlations were found with EDSS scores (manual: r = 0.337, p = 0.021; automatic: r = 0.346, p = 0.017). Meanwhile, in dataset1b, both manual and automatic segmentation revealed a significant increase in nCPV from baseline to follow-up (p < 0.05). The increase of nCPV was more pronounced in patients with disability worsened than stable patients (manual: p = 0.023; automatic: p = 0.018). Patients receiving disease-modifying therapy (DMT) exhibited a significantly lower nCPV increase than untreated patients (manual: p = 0.004; automatic: p = 0.004). CONCLUSION The 3D UX-Net model demonstrated strong segmentation performance for the CP, and the automatic segmented CP can be directly used in MS clinical practice. CP volume can serve as a surrogate imaging biomarker for monitoring disease progression and DMT response in MS patients.
Collapse
Affiliation(s)
- Xiaohua Wang
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China; Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaolong Wang
- College of Computer and Information Science, Southwest University, Chongqing, 400054, China
| | - Zichun Yan
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Feiyue Yin
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xiaojuan Liu
- College of Computer and Information Science, Southwest University, Chongqing, 400054, China.
| | - Yanbing Liu
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
20
|
Hemond CC, Gaitán MI, Absinta M, Reich DS. New Imaging Markers in Multiple Sclerosis and Related Disorders: Smoldering Inflammation and the Central Vein Sign. Neuroimaging Clin N Am 2024; 34:359-373. [PMID: 38942521 PMCID: PMC11213979 DOI: 10.1016/j.nic.2024.03.004] [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: 06/30/2024]
Abstract
Concepts of multiple sclerosis (MS) biology continue to evolve, with observations such as "progression independent of disease activity" challenging traditional phenotypic categorization. Iron-sensitive, susceptibility-based imaging techniques are emerging as highly translatable MR imaging sequences that allow for visualization of at least 2 clinically useful biomarkers: the central vein sign and the paramagnetic rim lesion (PRL). Both biomarkers demonstrate high specificity in the discrimination of MS from other mimics and can be seen at 1.5 T and 3 T field strengths. Additionally, PRLs represent a subset of chronic active lesions engaged in "smoldering" compartmentalized inflammation behind an intact blood-brain barrier.
Collapse
Affiliation(s)
- Christopher C Hemond
- Department of Neurology, University of Massachusetts Memorial Medical Center and University of Massachusetts Chan Medical School, Worcester, MA, USA; National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - María I Gaitán
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Martina Absinta
- Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
21
|
Jakimovski D, Zivadinov R, Qureshi F, Ramanathan M, Weinstock-Guttman B, Tavazzi E, Dwyer MG, Bergsland N. Serum Biomarker Signatures of Choroid Plexus Volume Changes in Multiple Sclerosis. Biomolecules 2024; 14:824. [PMID: 39062538 PMCID: PMC11275042 DOI: 10.3390/biom14070824] [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: 05/22/2024] [Revised: 06/18/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Increased choroid plexus (CP) volume has been recently implicated as a potential predictor of worse multiple sclerosis (MS) outcomes. The biomarker signature of CP changes in MS are currently unknown. To determine the blood-based biomarker characteristics of the cross-sectional and longitudinal MRI-based CP changes in a heterogeneous group of people with MS (pwMS), a total of 202 pwMS (148 pwRRMS and 54 pwPMS) underwent MRI examination at baseline and at a 5-year follow-up. The CP was automatically segmented and subsequently refined manually in order to obtain a normalized CP volume. Serum samples were collected at both timepoints, and the concentration of 21 protein measures relevant to MS pathophysiology were determined using the Olink™ platform. Age-, sex-, and BMI-adjusted linear regression models explored the cross-sectional and longitudinal relationships between MRI CP outcomes and blood-based biomarkers. At baseline, there were no significant proteomic predictors of CP volume, while at follow-up, greater CP volume was significantly associated with higher neurofilament light chain levels, NfL (standardized β = 0.373, p = 0.001), and lower osteopontin levels (standardized β = -0.23, p = 0.02). Higher baseline GFAP and lower FLRT2 levels were associated with future 5-year CP % volume expansion (standardized β = 0.277, p = 0.004 and standardized β = -0.226, p = 0.014, respectively). The CP volume in pwMS is associated with inflammatory blood-based biomarkers of neuronal injury (neurofilament light chain; NfL) and glial activation such as GFAP, osteopontin, and FLRT2. The expansion of the CP may play a central role in chronic and compartmentalized inflammation and may be driven by glial changes.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
- Wynn Hospital, Mohawk Valley Health System (MVHS), Utica, NY 13502, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
- Center for Biomedical Imaging at the Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | | | - Murali Ramanathan
- Department of Pharmaceutical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Bianca Weinstock-Guttman
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Eleonora Tavazzi
- Multiple Sclerosis Centre, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Michael G. Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| |
Collapse
|
22
|
Jiang J, Zhuo Z, Wang A, Li W, Jiang S, Duan Y, Ren Q, Zhao M, Wang L, Yang S, Awan MUN, Liu Y, Xu J. Choroid plexus volume as a novel candidate neuroimaging marker of the Alzheimer's continuum. Alzheimers Res Ther 2024; 16:149. [PMID: 38961406 PMCID: PMC11221040 DOI: 10.1186/s13195-024-01520-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Enlarged choroid plexus (ChP) volume has been reported in patients with Alzheimer's disease (AD) and inversely correlated with cognitive performance. However, its clinical diagnostic and predictive value, and mechanisms by which ChP impacts the AD continuum remain unclear. METHODS This prospective cohort study enrolled 607 participants [healthy control (HC): 110, mild cognitive impairment (MCI): 269, AD dementia: 228] from the Chinese Imaging, Biomarkers, and Lifestyle study between January 1, 2021, and December 31, 2022. Of the 497 patients on the AD continuum, 138 underwent lumbar puncture for cerebrospinal fluid (CSF) hallmark testing. The relationships between ChP volume and CSF pathological hallmarks (Aβ42, Aβ40, Aβ42/40, tTau, and pTau181), neuropsychological tests [Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), Neuropsychiatric Inventory (NPI), and Activities of Daily Living (ADL) scores], and multimodal neuroimaging measures [gray matter volume, cortical thickness, and corrected cerebral blood flow (cCBF)] were analyzed using partial Spearman's correlation. The mediating effects of four neuroimaging measures [ChP volume, hippocampal volume, lateral ventricular volume (LVV), and entorhinal cortical thickness (ECT)] on the relationship between CSF hallmarks and neuropsychological tests were examined. The ability of the four neuroimaging measures to identify cerebral Aβ42 changes or differentiate among patients with AD dementia, MCI and HCs was determined using receiver operating characteristic analysis, and their associations with neuropsychological test scores at baseline were evaluated by linear regression. Longitudinal associations between the rate of change in the four neuroimaging measures and neuropsychological tests scores were evaluated on the AD continuum using generalized linear mixed-effects models. RESULTS The participants' mean age was 65.99 ± 8.79 years. Patients with AD dementia exhibited the largest baseline ChP volume than the other groups (P < 0.05). ChP volume enlargement correlated with decreased Aβ42 and Aβ40 levels; lower MMSE and MoCA and higher NPI and ADL scores; and lower volume, cortical thickness, and cCBF in other cognition-related regions (all P < 0.05). ChP volume mediated the association of Aβ42 and Aβ40 levels with MMSE scores (19.08% and 36.57%), and Aβ42 levels mediated the association of ChP volume and MMSE or MoCA scores (39.49% and 34.36%). ChP volume alone better identified cerebral Aβ42 changes than LVV alone (AUC = 0.81 vs. 0.67, P = 0.04) and EC thickness alone (AUC = 0.81 vs.0.63, P = 0.01) and better differentiated patients with MCI from HCs than hippocampal volume alone (AUC = 0.85 vs. 0.81, P = 0.01), and LVV alone (AUC = 0.85 vs.0.82, P = 0.03). Combined ChP and hippocampal volumes significantly increased the ability to differentiate cerebral Aβ42 changes and patients among AD dementia, MCI, and HCs groups compared with hippocampal volume alone (all P < 0.05). After correcting for age, sex, years of education, APOE ε4 status, eTIV, and hippocampal volume, ChP volume was associated with MMSE, MoCA, NPI, and ADL score at baseline, and rapid ChP volume enlargement was associated with faster deterioration in NPI scores with an average follow-up of 10.03 ± 4.45 months (all P < 0.05). CONCLUSIONS ChP volume may be a novel neuroimaging marker associated with neurodegenerative changes and clinical AD manifestations. It could better detect the early stages of the AD and predict prognosis, and significantly enhance the differential diagnostic ability of hippocampus on the AD continuum.
Collapse
Affiliation(s)
- Jiwei Jiang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhizheng Zhuo
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Anxin Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wenyi Li
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shirui Jiang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yunyun Duan
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiwei Ren
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Min Zhao
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Linlin Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shiyi Yang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | | | - Yaou Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Jun Xu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
23
|
Bergsland N, Dwyer MG, Jakimovski D, Tavazzi E, Weinstock-Guttman B, Zivadinov R. Choroid plexus enlargement is associated with future periventricular neurodegeneration in multiple sclerosis. Mult Scler Relat Disord 2024; 87:105668. [PMID: 38744032 DOI: 10.1016/j.msard.2024.105668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/10/2024] [Accepted: 05/05/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND The choroid plexus (CP), located within the ventricles of the brain and the primary producer of cerebrospinal fluid, has been shown to be enlarged in patients with multiple sclerosis (MS) and linked to periventricular remyelination failure. Atrophied T2-lesion volume (aT2-LV), a promising neurodegenerative imaging marker in progressive MS (PMS), reflects the volume of periventricular lesions subsumed into cerebrospinal fluid over the follow-up. METHODS In a cohort of 143 people with relapsing-remitting MS (RRMS) and 53 with PMS, we used 3T magnetic resonance imaging (MRI) to quantify CP volume (CPV) at baseline and aT2-LV over an average of 5.4 years of follow-up. Partial correlations, adjusting for age and sex, and linear regression analyses were used to assess the relationships between imaging measures. RESULTS In both cohorts, CPV was associated with aT2-LV in both the RRMS group (r = 0.329, p < 0.001) as well as the PMS group (r = 0.522, p < 0.001). In regression analyses predicting aT2-LV, ventricular volume (final adjusted R2 = 0.407, p < 0.001) explained additional variance beyond age, sex, and T2-lesion volume in the RRMS group while CPV (final adjusted R2 = 0.446, p = 0.009) was retained in the PMS group. CONCLUSION Findings from this study suggest that the CP enlargement is associated with future neurodegeneration, with a particularly relevant role in PMS.
Collapse
Affiliation(s)
- Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Eleonora Tavazzi
- Multiple Sclerosis Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Bianca Weinstock-Guttman
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging at the Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
24
|
Levit E, Ren Z, Gonzenbach V, Azevedo CJ, Calabresi PA, Cree BA, Freeman L, Longbrake EE, Oh J, Schindler MK, Sicotte NL, Reich DS, Ontaneda D, Sati P, Cao Q, Shinohara RT, Solomon AJ. Choroid plexus volume differentiates MS from its mimics. Mult Scler 2024; 30:1072-1076. [PMID: 38481081 PMCID: PMC11288781 DOI: 10.1177/13524585241238094] [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: 07/31/2024]
Abstract
This study aimed to determine whether choroid plexus volume (CPV) could differentiate multiple sclerosis (MS) from its mimics. A secondary analysis of two previously enrolled studies, 50 participants with MS and 64 with alternative diagnoses were included. CPV was automatically segmented from 3T magnetic resonance imaging (MRI), followed by manual review to remove misclassified tissue. Mean normalized choroid plexus volume (nCPV) to intracranial volume demonstrated relatively high specificity for MS participants in each cohort (0.80 and 0.76) with an area under the receiver-operator characteristic curve of 0.71 (95% confidence interval (CI) = 0.55-0.87) and 0.65 (95% CI = 0.52-0.77). In this preliminary study, nCPV differentiated MS from its mimics.
Collapse
Affiliation(s)
- Elle Levit
- Department of Neurological Sciences, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
- The University of Vermont Medical Center, Burlington, VT, USA
| | - Zheng Ren
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology and Informatics and Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Virgilio Gonzenbach
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology and Informatics and Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christina J Azevedo
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peter A Calabresi
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bruce Ac Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Leorah Freeman
- Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | | | - Jiwon Oh
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Matthew K Schindler
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nancy L Sicotte
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH, USA
| | - Pascal Sati
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Quy Cao
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology and Informatics and Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Russell T Shinohara
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology and Informatics and Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrew J Solomon
- Department of Neurological Sciences, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
- The University of Vermont Medical Center, Burlington, VT, USA
| |
Collapse
|
25
|
Zivadinov R, Pei J, Clayton D, Goldman DE, Winger RC, Cabatingan MS, Dwyer MG, Bergsland N. Evolution of atrophied T2 lesion volume in primary-progressive multiple sclerosis: results from the phase 3 ORATORIO study. J Neurol Neurosurg Psychiatry 2024; 95:536-543. [PMID: 38071574 DOI: 10.1136/jnnp-2023-332573] [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: 09/05/2023] [Accepted: 11/20/2023] [Indexed: 12/22/2023]
Abstract
BACKGROUND Atrophied T2-lesion volume (aT2-LV) is an exploratory imaging marker in multiple sclerosis (MS) reflecting the volume of lesions subsumed into cerebrospinal fluid (CSF). OBJECTIVE To investigate the effect of ocrelizumab (OCR) versus placebo (PBO) over 120 weeks on the accumulation of aT2-LV in a double-blind placebo-controlled (DBP) phase 3, primary-progressive (PP) MS study (ORATORIO; NCT01194570). METHODS This post-hoc, MRI-blinded analysis evaluated 732 PPMS randomised to OCR (488) or PBO (244). Atrophied T2-LV was calculated by overlaying baseline T2-lesion masks on follow-up CSF maps. Clinical data from DBP and open-label extension (OLE) periods were available. Treatment effect was evaluated by a mixed-effect model with repeated measures, while logistic regression explored the association of aT2-LV at week 120 and clinical outcomes in the OLE period. RESULTS OCR treatment significantly reduced accumulation of aT2-LV compared with PBO (319.4 mm3 vs 366.1 mm3, p=0.015) at 120 weeks. OCR showed superiority over PBO in reducing aT2-LV in patients who developed confirmed disability progression (CDP) during the DBP period at 12 (CDP12) and 24 (CDP24) weeks for the composite of Expanded Disability Status Scale (EDSS), Nine-Hole Peg Test and Timed 25-Foot Walk test. Accumulation of aT2-LV at week 120 was related to CDP12-EDSS (p=0.018) and CDP24-EDSS (p=0.022) in the OLE for the patients who were treated by PBO in the DBP only. CONCLUSIONS OCR showed a significant effect of reducing the accumulation of aT2-LV in PPMS in the DBP period and was related to CDP-EDSS in OLE only in the PBO arm.
Collapse
Affiliation(s)
- Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| | | | | | | | | | | | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
26
|
Storelli L, Pagani E, Rubin M, Margoni M, Filippi M, Rocca MA. A Fully Automatic Method to Segment Choroid Plexuses in Multiple Sclerosis Using Conventional MRI Sequences. J Magn Reson Imaging 2024; 59:1643-1652. [PMID: 37530734 DOI: 10.1002/jmri.28937] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND Choroid plexus (CP) volume has been recently proposed as a proxy for brain neuroinflammation in multiple sclerosis (MS). PURPOSE To develop and validate a fast automatic method to segment CP using routinely acquired brain T1-weighted and FLAIR MRI. STUDY TYPE Retrospective. POPULATION Fifty-five MS patients (33 relapsing-remitting, 22 progressive; mean age = 46.8 ± 10.2 years; 31 women) and 60 healthy controls (HC; mean age = 36.1 ± 12.6 years, 33 women). FIELD STRENGTH/SEQUENCE 3D T2-weighted FLAIR and 3D T1-weighted gradient echo sequences at 3.0 T. ASSESSMENT Brain tissues were segmented on T1-weighted sequences and a Gaussian Mixture Model (GMM) was fitted to FLAIR image intensities obtained from the ventricle masks of the SIENAX. A second GMM was then applied on the thresholded and filtered ventricle mask. CP volumes were automatically determined and compared with those from manual segmentation by two raters (with 3 and 10 years' experience; reference standard). CP volumes from previously published automatic segmentation methods (freely available Freesurfer [FS] and FS-GMM) were also compared with reference standard. Expanded Disability Status Scale (EDSS) score was assessed within 3 days of MRI. Computational time was assessed for each automatic technique and manual segmentation. STATISTICAL TESTS Comparisons of CP volumes with reference standard were evaluated with Bland Altman analysis. Dice similarity coefficients (DSC) were computed to assess automatic CP segmentations. Volume differences between MS and HC for each method were assessed with t-tests and correlations of CP volumes with EDSS were assessed with Pearson's correlation coefficients (R). A P value <0.05 was considered statistically significant. RESULTS Compared to manual segmentation, the proposed method had the highest segmentation accuracy (mean DSC = 0.65 ± 0.06) compared to FS (mean DSC = 0.37 ± 0.08) and FS-GMM (0.58 ± 0.06). The percentage CP volume differences relative to manual segmentation were -0.1% ± 0.23, 4.6% ± 2.5, and -0.48% ± 2 for the proposed method, FS, and FS-GMM, respectively. The Pearson's correlations between automatically obtained CP volumes and the manually obtained volumes were 0.70, 0.54, and 0.56 for the proposed method, FS, and FS-GMM, respectively. A significant correlation between CP volume and EDSS was found for the proposed automatic pipeline (R = 0.2), for FS-GMM (R = 0.3) and for manual segmentation (R = 0.4). Computational time for the proposed method (32 ± 2 minutes) was similar to the manual segmentation (20 ± 5 minutes) but <25% of the FS (120 ± 15 minutes) and FS-GMM (125 ± 15 minutes) methods. DATA CONCLUSION This study developed an accurate and easily implementable method for automatic CP segmentation in MS using T1-weighted and FLAIR MRI. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 4.
Collapse
Affiliation(s)
- Loredana Storelli
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Pagani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Rubin
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Monica Margoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
27
|
Xie Y, Zhu H, Yao Y, Liu C, Wu S, Zhang Y, Zhu W. Enlarged choroid plexus in relapsing-remitting multiple sclerosis may lead to brain structural changes through the glymphatic impairment. Mult Scler Relat Disord 2024; 85:105550. [PMID: 38493535 DOI: 10.1016/j.msard.2024.105550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/22/2024] [Accepted: 03/10/2024] [Indexed: 03/19/2024]
Abstract
OBJECTIVES To investigate the potential link among choroid plexus (CP) volume, glymphatic clearance and brain structural change in relapsing-remitting multiple sclerosis (RRMS) patients. MATERIALS AND METHODS Sixty-five RRMS patients and 48 healthy controls (HC) underwent MRI examination. The diffusion tensor image analysis along the perivascular space (DTI-ALPS) was calculated to reflect glymphatic system function. The brain structure volume and DTI-ALPS index were compared between RRMS and HC. The mediating effect of the DTI-ALPS index between CP volume and brain structural changes was further investigated. The longitudinal changes of brain structure and DTI-ALPS index were compared in 20 RRMS patients. RESULTS Compared to HC, CP volume in RRMS was significantly increased (P < 0.001), and DTI-ALPS index was significantly decreased (P = 0.001). The volumes of white matter, thalamus, putamen and pallidum were significantly decreased in RRMS, and the volumes of lateral ventricle and third ventricle were increased. Mediation analysis showed DTI-ALPS index partially mediated the association between CP enlargement and deep gray matter (DGM) atrophy in RRMS, and between CP enlargement and ventricle enlargement. CP volume and DTI-ALPS index were also significantly correlated with Expanded Disability Status Scale (EDSS) (P = 0.006, P = 0.043). Notably, the variation of DTI_ALPS index during the follow-up period were significantly and negatively correlated with the variation of EDSS (P = 0.045). CONCLUSION Enlarged CP volume and decreased DTI_ALPS index may be closely related to DGM atrophy and ventricular enlargement in RRMS, and may be potential imaging markers of clinical disability.
Collapse
Affiliation(s)
- Yan Xie
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Hongquan Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yihao Yao
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Chengxia Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Shaolong Wu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
28
|
Manelis A, Hu H, Miceli R, Satz S, Lau R, Iyengar S, Swartz HA. The relationship between the size and asymmetry of the lateral ventricles and cortical myelin content in individuals with mood disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.30.24306621. [PMID: 38746112 PMCID: PMC11092679 DOI: 10.1101/2024.04.30.24306621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Although enlargement of the lateral ventricles was previously observed in individuals with mood disorders, the link between ventricular size and asymmetry with other indices of brain structure remains underexplored. In this study, we examined the association of lateral ventricular size and asymmetry with cortical myelin content in individuals with bipolar (BD) and depressive (DD) disorders compared to healthy controls (HC). Methods Magnetic resonance imaging (MRI) was used to obtain T1w and T2w images from 149 individuals (age=27.7 (SD=6.1) years, 78% female, BD=38, DD=57, HC=54). Cortical myelin content was calculated using the T1w/T2w ratio. Elastic net regularized regression identified brain regions whose myelin content was associated with ventricular size and asymmetry. A post-hoc linear regression examined how participants' diagnosis, illness duration, and current level of depression moderated the relationship between the size and asymmetry of the lateral ventricles and levels of cortical myelin in the selected brain regions. Results Individuals with mood disorders had larger lateral ventricles than HC. Larger ventricles and lower asymmetry were observed in individuals with BD who had longer lifetime illness duration and more severe current depressive symptoms. A greater left asymmetry was observed in participants with DD than in those with BD (p<0.01). Elastic net revealed that both ventricular enlargement and asymmetry were associated with altered myelin content in cingulate, frontal, and sensorimotor cortices. In BD, but not other groups, ventricular enlargement was related to altered myelin content in the right insular regions. Conclusions Lateral ventricular enlargement and asymmetry are linked to myelin content imbalance, thus, potentially leading to emotional and cognitive dysfunction in mood disorders.
Collapse
|
29
|
Andravizou A, Stavropoulou De Lorenzo S, Kesidou E, Michailidou I, Parissis D, Boziki MK, Stamati P, Bakirtzis C, Grigoriadis N. The Time Trajectory of Choroid Plexus Enlargement in Multiple Sclerosis. Healthcare (Basel) 2024; 12:768. [PMID: 38610190 PMCID: PMC11011748 DOI: 10.3390/healthcare12070768] [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: 02/27/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Choroid plexus (CP) can be seen as a watchtower of the central nervous system (CNS) that actively regulates CNS homeostasis. A growing body of literature suggests that CP alterations are involved in the pathogenesis of multiple sclerosis (MS) but the underlying mechanisms remain elusive. CPs are enlarged and inflamed in relapsing-remitting (RRMS) but also in clinically isolated syndrome (CIS) and radiologically isolated syndrome (RIS) stages, far beyond MS diagnosis. Increases in the choroid plexus/total intracranial volume (CP/TIV) ratio have been robustly associated with increased lesion load, higher translocator protein (TSPO) uptake in normal-appearing white matter (NAWM) and thalami, as well as with higher annual relapse rate and disability progression in highly active RRMS individuals, but not in progressive MS. The CP/TIV ratio has only slightly been correlated with magnetic resonance imaging (MRI) findings (cortical or whole brain atrophy) and clinical outcomes (EDSS score) in progressive MS. Therefore, we suggest that plexus volumetric assessments should be mainly applied to the early disease stages of MS, whereas it should be taken into consideration with caution in progressive MS. In this review, we attempt to clarify the pathological significance of the temporal CP volume (CPV) changes in MS and highlight the pitfalls and limitations of CP volumetric analysis.
Collapse
Affiliation(s)
- Athina Andravizou
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Sotiria Stavropoulou De Lorenzo
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Evangelia Kesidou
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Iliana Michailidou
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Dimitrios Parissis
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Marina-Kleopatra Boziki
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Polyxeni Stamati
- Department of Neurology, University Hospital of Larissa, 41334 Larissa, Greece;
| | - Christos Bakirtzis
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Nikolaos Grigoriadis
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| |
Collapse
|
30
|
Klistorner S, Barnett MH, Wang C, Parratt J, Yiannikas C, Klistorner A. Longitudinal enlargement of choroid plexus is associated with chronic lesion expansion and neurodegeneration in RRMS patients. Mult Scler 2024; 30:496-504. [PMID: 38318807 PMCID: PMC11010552 DOI: 10.1177/13524585241228423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/27/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND AND OBJECTIVE We explored dynamic changes in the choroid plexus (CP) in patients with relapsing-remitting multiple sclerosis (RRMS) and assessed its relationship with chronic lesion expansion and atrophy in various brain compartments. METHODS Fifty-seven RRMS patients were annually assessed for a minimum of 48 months with 3D FLAIR, pre- and post-contrast 3D T1 and diffusion-weighted magnetic resonance imaging (MRI). The CP was manually segmented at baseline and last follow-up. RESULTS The volume of CP significantly increased by 1.4% annually. However, the extent of CP enlargement varied considerably among individuals (ranging from -3.6 to 150.8 mm3 or -0.2% to 6.3%). The magnitude of CP enlargement significantly correlated with central (r = 0.70, p < 0.001) and total brain atrophy (r = -0.57, p < 0.001), white (r = -0.61, p < 0.001) and deep grey matter atrophy (r = -0.60, p < 0.001). Progressive CP enlargement was significantly associated with the volume and extent of chronic lesion expansion (r = 0.60, p < 0.001), but not with the number or volume of new lesions. CONCLUSION This study provides evidence of progressive CP enlargement in patients with RRMS. Our findings also demonstrate that enlargement of the CP volume is linked to the expansion of chronic lesions and neurodegeneration of periventricular white and grey matter in RRMS patients.
Collapse
Affiliation(s)
- Samuel Klistorner
- Save Sight Institute, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Michael H Barnett
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Centre, Camperdown, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Chenyu Wang
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia/Sydney Neuroimaging Analysis Centre, Camperdown, NSW, Australia
| | - John Parratt
- Royal North Shore Hospital, Sydney, NSW, Australia
| | | | - Alexander Klistorner
- Save Sight Institute, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
31
|
Ueno M, Chiba Y, Murakami R, Miyai Y, Matsumoto K, Wakamatsu K, Nakagawa T, Takebayashi G, Uemura N, Yanase K, Ogino Y. Transporters, Ion Channels, and Junctional Proteins in Choroid Plexus Epithelial Cells. Biomedicines 2024; 12:708. [PMID: 38672064 PMCID: PMC11048166 DOI: 10.3390/biomedicines12040708] [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: 02/24/2024] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
The choroid plexus (CP) plays significant roles in secreting cerebrospinal fluid (CSF) and forming circadian rhythms. A monolayer of epithelial cells with tight and adherens junctions of CP forms the blood-CSF barrier to control the movement of substances between the blood and ventricles, as microvessels in the stroma of CP have fenestrations in endothelial cells. CP epithelial cells are equipped with several kinds of transporters and ion channels to transport nutrient substances and secrete CSF. In addition, junctional components also contribute to CSF production as well as blood-CSF barrier formation. However, it remains unclear how junctional components as well as transporters and ion channels contribute to the pathogenesis of neurodegenerative disorders. In this manuscript, recent findings regarding the distribution and significance of transporters, ion channels, and junctional proteins in CP epithelial cells are introduced, and how changes in expression of their epithelial proteins contribute to the pathophysiology of brain disorders are reviewed.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Ryuta Murakami
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Yumi Miyai
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Koichi Matsumoto
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Keiji Wakamatsu
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Toshitaka Nakagawa
- Division of Research Instrument and Equipment, Research Facility Center, Kagawa University, Kagawa 761-0793, Japan;
| | - Genta Takebayashi
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| | - Naoya Uemura
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| | - Ken Yanase
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| | - Yuichi Ogino
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| |
Collapse
|
32
|
Preziosa P, Pagani E, Meani A, Storelli L, Margoni M, Yudin Y, Tedone N, Biondi D, Rubin M, Rocca MA, Filippi M. Chronic Active Lesions and Larger Choroid Plexus Explain Cognition and Fatigue in Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200205. [PMID: 38350048 DOI: 10.1212/nxi.0000000000200205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/18/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND AND OBJECTIVES Chronic inflammation may contribute to cognitive dysfunction and fatigue in patients with multiple sclerosis (MS). Paramagnetic rim lesions (PRLs) and choroid plexus (CP) enlargement have been proposed as markers of chronic inflammation in MS being associated with a more severe disease course. However, their relation with cognitive impairment and fatigue has not been fully explored yet. Here, we investigated the contribution of PRL number and volume and CP enlargement to cognitive impairment and fatigue in patients with MS. METHODS Brain 3T MRI, neurologic evaluation, and neuropsychological assessment, including the Brief Repeatable Battery of Neuropsychological Tests and Modified Fatigue Impact Scale, were obtained from 129 patients with MS and 73 age-matched and sex-matched healthy controls (HC). PRLs were identified on phase images of susceptibility-weighted imaging, whereas CP volume was quantified using a fully automatic method on brain three-dimensional T1-weighted and fluid-attenuated inversion recovery MRI sequences. Predictors of cognitive impairment and fatigue were identified using random forest. RESULTS Thirty-six (27.9%) patients with MS were cognitively impaired, and 31/113 (27.4%) patients had fatigue. Fifty-nine (45.7%) patients with MS had ≥1 PRLs (median = 0, interquartile range = 0;2). Compared with HC, patients with MS showed significantly higher T2-hyperintense white matter lesion (WM) volume; lower normalized brain, thalamic, hippocampal, caudate, cortical, and WM volumes; and higher normalized CP volume (p from <0.001 to 0.040). The predictors of cognitive impairment (relative importance) (out-of-bag area under the curve [OOB-AUC] = 0.707) were normalized brain volume (100%), normalized caudate volume (89.1%), normalized CP volume (80.3%), normalized cortical volume (70.3%), number (67.3%) and volume (66.7%) of PRLs, and T2-hyperintense WM lesion volume (64.0%). Normalized CP volume was the only predictor of the presence of fatigue (OOB-AUC = 0.563). DISCUSSION Chronic inflammation, with higher number and volume of PRLs and enlarged CP, may contribute to cognitive impairment in MS in addition to gray matter atrophy. The contribution of enlarged CP in explaining fatigue supports the relevance of immune-related processes in determining this manifestation independently of disease severity. PRLs and CP enlargement may contribute to the pathophysiology of cognitive impairment and fatigue in MS, and they may represent clinically relevant therapeutic targets to limit the impact of these clinical manifestations in MS.
Collapse
Affiliation(s)
- Paolo Preziosa
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Pagani
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Meani
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Loredana Storelli
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Monica Margoni
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Yury Yudin
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicolò Tedone
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Diana Biondi
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Rubin
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria A Rocca
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
33
|
Petzold J, Pochon JBF, Ghahremani DG, London ED. Structural indices of brain aging in methamphetamine use disorder. Drug Alcohol Depend 2024; 256:111107. [PMID: 38330525 DOI: 10.1016/j.drugalcdep.2024.111107] [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: 08/22/2023] [Revised: 01/01/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Methamphetamine use is surging globally. It has been linked to premature stroke, Parkinsonism, and dementia, suggesting that it may accelerate brain aging. METHODS We performed a retrospective study to determine if structural indices of brain aging were more prevalent prior to old age (26 - 54 years) in individuals with Methamphetamine Use Disorder (MUD), who were in early abstinence (M ± SD = 22.1 ± 25.6 days) than in healthy control (HC) participants. We compared T1-weighted MRI brain scans in age- and sex-matched groups (n = 89/group) on three structural features of brain aging: the brain volume/cerebrospinal fluid (BV/CSF) index, volume of white matter hypointensities/lesions, and choroid plexus volume. RESULTS The MUD group had a lower mean BV/CSF index and larger volumes of white matter hypointensities and choroid plexus (p-values < 0.01). Regression analyses showed significant age-by-group effects, indicating different age trajectories of the BV/CSF index and choroid plexus volume, consistent with abnormal global brain atrophy and choroid plexus pathology in the MUD group. Significant age and group main effects reflected a larger volume of white matter hypointensities for older participants across groups and for the MUD group irrespective of age. None of the three measures of brain aging correlated significantly with recent use or duration of recent abstinence from methamphetamine. CONCLUSIONS Premature brain pathology, which may reflect cerebrovascular damage and dysfunction of the choroid plexus, occurs in people with MUD. Such pathology may affect cognition and thereby efficacy of behavioral treatments for MUD.
Collapse
Affiliation(s)
- Johannes Petzold
- Jane and Terry Semel Institute of Neuroscience and Human Behavior, and Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Psychotherapy, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jean-Baptiste F Pochon
- Jane and Terry Semel Institute of Neuroscience and Human Behavior, and Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
| | - Dara G Ghahremani
- Jane and Terry Semel Institute of Neuroscience and Human Behavior, and Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
| | - Edythe D London
- Jane and Terry Semel Institute of Neuroscience and Human Behavior, and Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA; The Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
34
|
Visani V, Pizzini FB, Natale V, Tamanti A, Anglani M, Bertoldo A, Calabrese M, Castellaro M. Choroid plexus volume in multiple sclerosis can be estimated on structural MRI avoiding contrast injection. Eur Radiol Exp 2024; 8:33. [PMID: 38409562 PMCID: PMC10897123 DOI: 10.1186/s41747-024-00421-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/11/2023] [Indexed: 02/28/2024] Open
Abstract
We compared choroid plexus (ChP) manual segmentation on non-contrast-enhanced (non-CE) sequences and reference standard CE T1- weighted (T1w) sequences in 61 multiple sclerosis patients prospectively included. ChP was separately segmented on T1w, T2-weighted (T2w) fluid-attenuated inversion-recovery (FLAIR), and CE-T1w sequences. Inter-rater variability assessed on 10 subjects showed high reproducibility between sequences measured by intraclass correlation coefficient (T1w 0.93, FLAIR 0.93, CE-T1w 0.99). CE-T1w showed higher signal-to-noise ratio and contrast-to-noise ratio (CE-T1w 23.77 and 18.49, T1w 13.73 and 7.44, FLAIR 13.09 and 10.77, respectively). Manual segmentation of ChP resulted 3.073 ± 0.563 mL (mean ± standard deviation) on T1w, 3.787 ± 0.679 mL on FLAIR, and 2.984 ± 0.506 mL on CE-T1w images, with an error of 28.02 ± 19.02% for FLAIR and 3.52 ± 12.61% for T1w. FLAIR overestimated ChP volume compared to CE-T1w (p < 0.001). The Dice similarity coefficient of CE-T1w versus T1w and FLAIR was 0.67 ± 0.05 and 0.68 ± 0.05, respectively. Spatial error distribution per slice was calculated after nonlinear coregistration to the standard MNI152 space and showed a heterogeneous profile along the ChP especially near the fornix and the hippocampus. Quantitative analyses suggest T1w as a surrogate of CE-T1w to estimate ChP volume.Relevance statement To estimate the ChP volume, CE-T1w can be replaced by non-CE T1w sequences because the error is acceptable, while FLAIR overestimates the ChP volume. This encourages the development of automatic tools for ChP segmentation, also improving the understanding of the role of the ChP volume in multiple sclerosis, promoting longitudinal studies.Key points • CE-T1w sequences are considered the reference standard for ChP manual segmentation.• FLAIR sequences showed a higher CNR than T1w sequences but overestimated the ChP volume.• Non-CE T1w sequences can be a surrogate of CE-T1w sequences for manual segmentation of ChP.
Collapse
Affiliation(s)
- Valentina Visani
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Francesca B Pizzini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Valerio Natale
- Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Agnese Tamanti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Alessandra Bertoldo
- Department of Information Engineering, University of Padova, Padova, Italy
- Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Massimiliano Calabrese
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marco Castellaro
- Department of Information Engineering, University of Padova, Padova, Italy.
| |
Collapse
|
35
|
Raghib MF, Bao F, Elkhooly M, Bernitsas E. Choroid plexus volume as a marker of retinal atrophy in relapsing remitting multiple sclerosis. J Neurol Sci 2024; 457:122884. [PMID: 38237367 DOI: 10.1016/j.jns.2024.122884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 02/17/2024]
Abstract
OBJECTIVE To evaluate choroid plexus (CP) volume as a biomarker for predicting clinical disability and retinal layer atrophy in relapsing remitting multiple sclerosis (RRMS). METHODS Ninety-five RRMS patients and 26 healthy controls (HCs) underwent 3 T whole brain MRI, expanded disability status scale (EDSS) and optical coherence tomography (OCT). Fully automated intra-retinal segmentation was performed to obtain the volumes of the retinal nerve fiber layer (RNFL), combined ganglion cell layer -inner plexiform layer (GCIPL), inner nuclear layer (INL), outer plexiform layer (OPL), outer nuclear layer (ONL), retinal pigment epithelium (RPE), total macular volume (TMV) and papillomacular bundle (PMB). Automated segmentation of the CP within the lateral ventricles was performed and the choroid plexus volume (CPV) was normalized by total intracranial volume (TIV). Linear regression analysis and generalized estimating equation (GEE) models were applied to evaluate relationships between nCPV and EDSS, T2 lesion volume, disease duration, and retinal layer volumes, followed by Bonferroni correction analysis for multiple comparisons. RESULTS RRMS patients had larger tChPV compared to HCs (p < 0.001). After Bonferroni correction, there was a significant positive correlation between tChPV and EDSS (r2 = 0.25, p = 0.0002), disease duration (r2 = 0.30, p = 0.01), and T2 lesion volume (r2 = 0.39, p = 0.0000). A robust negative correlation was found between tChPV and RNFL (p < 0.001), GCIPL (p = 0.003), TMV (p = 0.0185), PMB (p < 0.0001), G (p = 0.04), T(p = 0.0001). CONCLUSIONS Our findings support the association of tChPV with disability and altered retinal integrity in RRMS.
Collapse
Affiliation(s)
- Muhammad F Raghib
- Department of Neurology, Wayne State University School of Medicine, United States of America
| | - Fen Bao
- Department of Neurology, Wayne State University School of Medicine, United States of America
| | - Mahmoud Elkhooly
- Department of Neurology, Wayne State University School of Medicine, United States of America; Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, United States of America; Department of Neurology and Psychiatry, Minia University, Minia, Egypt
| | - Evanthia Bernitsas
- Department of Neurology, Wayne State University School of Medicine, United States of America; Detroit Medical Center, Detroit, MI, United States of America.
| |
Collapse
|
36
|
Murck H, Karailiev P, Karailievova L, Puhova A, Jezova D. Treatment with Glycyrrhiza glabra Extract Induces Anxiolytic Effects Associated with Reduced Salt Preference and Changes in Barrier Protein Gene Expression. Nutrients 2024; 16:515. [PMID: 38398838 PMCID: PMC10893552 DOI: 10.3390/nu16040515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
We have previously identified that low responsiveness to antidepressive therapy is associated with higher aldosterone/cortisol ratio, lower systolic blood pressure, and higher salt preference. Glycyrrhiza glabra (GG) contains glycyrrhizin, an inhibitor of 11β-hydroxysteroid-dehydrogenase type-2 and antagonist of toll-like receptor 4. The primary hypothesis of this study is that food enrichment with GG extract results in decreased anxiety behavior and reduced salt preference under stress and non-stress conditions. The secondary hypothesis is that the mentioned changes are associated with altered gene expression of barrier proteins in the prefrontal cortex. Male Sprague-Dawley rats were exposed to chronic mild stress for five weeks. Both stressed and unstressed rats were fed a diet with or without an extract of GG roots for the last two weeks. GG induced anxiolytic effects in animals independent of stress exposure, as measured in elevated plus maze test. Salt preference and intake were significantly reduced by GG under control, but not stress conditions. The gene expression of the barrier protein claudin-11 in the prefrontal cortex was increased in control rats exposed to GG, whereas stress-induced rise was prevented. Exposure to GG-enriched diet resulted in reduced ZO-1 expression irrespective of stress conditions. In conclusion, the observed effects of GG are in line with a reduction in the activity of central mineralocorticoid receptors. The treatment with GG extract or its active components may, therefore, be a useful adjunct therapy for patients with subtypes of depression and anxiety disorders with heightened renin-angiotensin-aldosterone system and/or inflammatory activity.
Collapse
Affiliation(s)
- Harald Murck
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, 35039 Marburg, Germany
| | - Peter Karailiev
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Lucia Karailievova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Agnesa Puhova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| |
Collapse
|
37
|
Murck H, Fava M, Cusin C, Fatt CC, Trivedi M. Brain ventricle and choroid plexus morphology as predictor of treatment response in major depression: Findings from the EMBARC study. Brain Behav Immun Health 2024; 35:100717. [PMID: 38186634 PMCID: PMC10767278 DOI: 10.1016/j.bbih.2023.100717] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/09/2024] Open
Abstract
Recent observations suggest a role of the volume of the cerebral ventricle volume, corpus callosum (CC) segment volume, in particular that of the central-anterior part, and choroid plexus (CP) volume for treatment resistance of major depressive disorder (MDD). An increased CP volume has been associated with increased inflammatory activity and changes in the structure of the ventricles and corpus callosum. We attempt to replicate and confirm that these imaging markers are associated with clinical outcome in subjects from the EMBARC study, as implied by a recent pilot study. The EMBARC study is a placebo controlled randomized study comparing sertraline vs. placebo in patients with MDD to identify biological markers of therapy resistance. Association of baseline volumes of the lateral ventricles (LVV), choroid plexus volume (CPV) and volume of segments of the CC with treatment response after 4 weeks treatment was evaluated. 171 subjects (61 male, 110 female) completed the 4 week assessments; gender and age were taken into account for this analyses. As previously reported, no treatment effect of sertraline vs. placebo was observed, therefore the study characterized prognostic markers of response in the pooled population. Change in depression severity was identified by the ratio of the Hamilton-Depression rating scale 17 (HAMD-17) at week 4 divided by the HAMD-17 at baseline (HAMD-17 ratio). Volumes of the lateral ventricles and choroid plexi were positively correlated with the HAMD-17 ratio, indication worse outcome with larger ventricles and choroid plexus volumes, whereas the volume of the central-anterior corpus callosum was negatively correlated with the HAMD-17 ratio. Responders (n = 54) had significantly smaller volumes of the lateral ventricles and CP compared to non-responders (n = 117), whereas the volume of mid-anterior CC was significantly larger compared to non-responders (n = 117), confirming our previous findings. In an exploratory way associations between enlarged LVV and CPV and signs of lipid dysregulation were observed. In conclusion, we confirmed that volumes of lateral ventricles, choroid plexi and the mid-anterior corpus callosum are associated with clinical improvement of depression and may be indicators of metabolic/inflammatory activity.
Collapse
Affiliation(s)
- Harald Murck
- Dept. of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Cusin
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cherise Chin Fatt
- The University of Texas Southwestern Medical Center, Department of Psychiatry, Center for Depression Research and Clinical Care, Dallas, USA
| | - Madhukar Trivedi
- The University of Texas Southwestern Medical Center, Department of Psychiatry, Center for Depression Research and Clinical Care, Dallas, USA
| |
Collapse
|
38
|
Akaishi T, Fujimori J, Nakashima I. Enlarged choroid plexus in multiple sclerosis is associated with increased lesion load and atrophy in white matter but not gray matter atrophy. Mult Scler Relat Disord 2024; 82:105424. [PMID: 38181695 DOI: 10.1016/j.msard.2024.105424] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/16/2023] [Accepted: 01/01/2024] [Indexed: 01/07/2024]
Abstract
BACKGROUND Enlargement of the choroid plexus (CP) is reported to associate with inflammatory activity and contribute to brain atrophy in patients with multiple sclerosis (pwMS). However, a recent study in healthy volunteers (HVTs) has suggested that CP enlargement can be attributed to ventriculomegaly. OBJECTIVES To clarify the pathological significance of the enlargement of CP in multiple sclerosis (MS). METHODS A total of 102 pwMS (89 with relapsing-remitting MS and 13 with secondary progressive MS) and 41 HVTs were cross-sectionally evaluated using brain volumetry. The CP volume was compared between disease groups and investigated for the relationships with other brain regional volumes. RESULTS CP volume was significantly larger in pwMS than in HVTs in the univariate analysis, but not in multivariable analysis. Meanwhile, the CP and lateral ventricle (LV) volumes were significantly correlated. CP enlargement was significantly associated with increased lesion load and cerebral white matter (WM) atrophy, even after adjusting for LV volume. In contrast, multivariable analyses revealed that LV enlargement, but not CP enlargement, was associated with total gray matter (GM) atrophy. CONCLUSION CP enlargement was closely associated with LV enlargement. After adjusting for LV volume, CP enlargement in pwMS was associated with increased lesion load and WM atrophy but not GM atrophy.
Collapse
Affiliation(s)
- Tetsuya Akaishi
- Department of Neurology, Tohoku University, Sendai, Japan; Department of Education and Support for Regional Medicine, Tohoku University, Sendai, Japan
| | - Juichi Fujimori
- Division of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
| | - Ichiro Nakashima
- Division of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
39
|
Bravi B, Melloni EMT, Paolini M, Palladini M, Calesella F, Servidio L, Agnoletto E, Poletti S, Lorenzi C, Colombo C, Benedetti F. Choroid plexus volume is increased in mood disorders and associates with circulating inflammatory cytokines. Brain Behav Immun 2024; 116:52-61. [PMID: 38030049 DOI: 10.1016/j.bbi.2023.11.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/17/2023] [Accepted: 11/26/2023] [Indexed: 12/01/2023] Open
Abstract
Depressed patients exhibit altered levels of immune-inflammatory markers both in the peripheral blood and in the cerebrospinal fluid (CSF) and inflammatory processes have been widely implicated in the pathophysiology of mood disorders. The Choroid Plexus (ChP), located at the base of each of the four brain ventricles, regulates the exchange of substances between the blood and CSF and several evidence supported a key role for ChP as a neuro-immunological interface between the brain and circulating immune cells. Given the role of ChP as a regulatory gate between periphery, CSF spaces and the brain, we compared ChP volumes in patients with bipolar disorder (BP) or major depressive disorder (MDD) and healthy controls, exploring their association with history of illness and levels of circulating cytokines. Plasma levels of inflammatory markers and MRI scans were acquired for 73 MDD, 79 BD and 72 age- and sex-matched healthy controls (HC). Patients with either BD or MDD had higher ChP volumes than HC. With increasing age, the bilateral ChP volume was larger in patients, an effect driven by the duration of illness; while only minor effects were observed in HC. Right ChP volumes were proportional to higher levels of circulating cytokines in the clinical groups, including IFN-γ, IL-13 and IL-17. Specific effects in the two diagnostic groups were observed when considering the left ChP, with positive association with IL-1ra, IL-13, IL-17, and CCL3 in BD, and negative associations with IL-2, IL-4, IL-1ra, and IFN-γ in MDD. These results suggest that ChP could represent a reliable and easy-to-assess biomarker to evaluate the brain effects of inflammatory status in mood disorders, contributing to personalized diagnosis and tailored treatment strategies.
Collapse
Affiliation(s)
- Beatrice Bravi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; PhD Program in Cognitive Neuroscience, University Vita-Salute San Raffaele, Milan, Italy.
| | - Elisa Maria Teresa Melloni
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Marco Paolini
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; PhD Program in Molecular Medicine, University Vita-Salute San Raffaele, Milan, Italy
| | - Mariagrazia Palladini
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; PhD Program in Cognitive Neuroscience, University Vita-Salute San Raffaele, Milan, Italy
| | - Federico Calesella
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; PhD Program in Cognitive Neuroscience, University Vita-Salute San Raffaele, Milan, Italy
| | - Laura Servidio
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Elena Agnoletto
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Sara Poletti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Cristina Lorenzi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Cristina Colombo
- University Vita-Salute San Raffaele, Milan, Italy; Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
40
|
Gueye M, Preziosa P, Ramirez GA, Bozzolo EP, Canti V, Margoni M, Meani A, Moiola L, Rovere-Querini P, Manfredi AA, Filippi M, Rocca MA. Choroid plexus and perivascular space enlargement in neuropsychiatric systemic lupus erythematosus. Mol Psychiatry 2024; 29:359-368. [PMID: 38036603 DOI: 10.1038/s41380-023-02332-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023]
Abstract
Choroid plexus (CP) enlargement is proposed as a marker of neuroinflammation in immune-mediated conditions. CP involvement has also been hypothesized in the immunopathology of systemic lupus erythematosus (SLE). We investigated whether CP enlargement occurs in SLE patients and its association with neuropsychiatric involvement. Additionally, we explored abnormalities along the glymphatic system in SLE patients through enlarged perivascular space (PVS) quantification. Clinical assessment and 3 Tesla brain dual-echo and T1-weighted MRI scans were obtained from 32 SLE patients and 32 sex and age-matched healthy controls (HC). CPs were manually segmented on 3D T1-weighted sequence and enlarged PVS (ePVS) were assessed through Potter's score. Compared to HC, SLE patients showed higher normalized CP volume (nCPV) (p = 0.023), with higher CP enlargement in neuropsychiatric SLE (NPSLE) (n = 12) vs. non-NPSLE (p = 0.027) patients. SLE patients with antiphospholipid antibodies (APA) positivity (n = 18) had higher nCPV compared to HC (p = 0.012), while APA negative ones did not. SLE patients also had higher Potter's score than HC (p < 0.001), with a tendency towards a higher number of basal ganglia ePVS in NPSLE vs. non-NPSLE patients. Using a random forest analysis, nCPV emerged as a significant predictor of NPSLE, together with T2-hyperintense white matter (WM) lesion volume (LV) and APA positivity (out-of-bag AUC 0.81). Our findings support the hypothesis of a role exerted by the CP in SLE physiopathology, especially in patients with neuropsychiatric involvement. The higher prevalence of ePVS in SLE patients, compared to HC, suggests the presence of glymphatic system impairment in this population.
Collapse
Affiliation(s)
- Mor Gueye
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Giuseppe A Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases & Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Enrica P Bozzolo
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Canti
- Unit of Internal Medicine & Division of Immunology, Transplantation and Infectious diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Monica Margoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Meani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Patrizia Rovere-Querini
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Internal Medicine & Division of Immunology, Transplantation and Infectious diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Angelo A Manfredi
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases & Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
41
|
Morozumi T, Preziosa P, Meani A, Albergoni M, Margoni M, Pagani E, Filippi M, Rocca MA. Influence of cardiorespiratory fitness and MRI measures of neuroinflammation on hippocampal volume in multiple sclerosis. J Neurol Neurosurg Psychiatry 2023; 95:29-36. [PMID: 37468307 DOI: 10.1136/jnnp-2023-331482] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND The hippocampus is a clinically relevant region where neurogenesis and neuroplasticity occur throughout the whole lifespan. Neuroinflammation and cardiorespiratory fitness (CRF) may influence hippocampal integrity by modulating the processes promoting neurogenesis and neuroprotection that contribute to the preservation of functions. This study aimed to investigate the effects of neuroinflammation and CRF on hippocampal volume in multiple sclerosis (MS) patients with relapsing-remitting (RR) and progressive (P) clinical phenotypes. The influence of neuroinflammation and CRF on brain, grey matter (GM) and thalamic volumes was also assessed to determine whether the effects were specific for the hippocampus. METHOD Brain 3T structural MRI scans and maximum oxygen consumption (VO2max), a proxy of CRF, were acquired from 81 MS patients (27 RR and 54 P) and 45 age-matched and sex-matched healthy controls. T2-hyperintense white matter lesion volume (T2-LV) and choroid plexuses volume (CPV) were quantified as neuroinflammatory measures. Associations of demographic, clinical, neuroinflammatory and CRF measures with normalised brain, GM, hippocampal and thalamic volumes in relapsing-remitting MS (RRMS) and progressive MS patients were assessed using Shapley and best subset selection regression. RESULTS For most volumetric measures, the largest portions of variance were explained by T2-LV (variable importance (VI)=9.4-39.4) and CPV (VI=4.5-26.2). VO2max explained the largest portion of variance of normalised hippocampal volume only in RRMS patients (VI=16.9) and was retained as relevant predictor (standardised β=0.374, p=0.023) with T2-LV (standardised β=-0.330, p=0.016). CONCLUSIONS A higher CRF may play a specific neuroprotective role on MS patients' hippocampal integrity, but only in the RR phase of the disease.
Collapse
Affiliation(s)
- Tetsu Morozumi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
- Neurology Unit, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Alessandro Meani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Matteo Albergoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Monica Margoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Elisabetta Pagani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
- Neurology Unit, IRCCS Ospedale San Raffaele, Milano, Italy
- Neurorehabilitation Unit, IRCCS Ospedale San Raffaele, Milano, Italy
- Neurophysiology Service, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
- Neurology Unit, IRCCS Ospedale San Raffaele, Milano, Italy
| |
Collapse
|
42
|
Tu Y, Li Z, Xiong F, Gao F. Decreased DTI-ALPS and choroid plexus enlargement in fibromyalgia: a preliminary multimodal MRI study. Neuroradiology 2023; 65:1749-1755. [PMID: 37870589 DOI: 10.1007/s00234-023-03240-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/09/2023] [Indexed: 10/24/2023]
Abstract
PURPOSE The glymphatic system is a fluid exchange pathway that clears waste products that is crucial for the maintenance of brain homeostasis. However, the exact role it plays in the emergence of fibromyalgia (FM) is still not fully understood. Here, we explored the changes in non-invasive MRI proxy probably related to the glymphatic function in FM patients, and explored brain-behavior relationships. METHODS A total of 40 participants, consisting of 20 individuals with FM and 20 healthy controls (HCs), were included in the study. The participants underwent structural T1-weighted MRI, diffusion tensor imaging (DTI), and clinical assessment. The data was obtained from an open access dataset. The study compared non-invasive MRI indices, including choroid plexus (CP) volume and DTI analysis along the perivascular space (ALPS), between the FM and HC groups. Furthermore, correlation analysis was conducted to determine the correlation between clinical parameters and both CP volume and DTI-ALPS index. RESULTS Patients with FM had significantly higher CP volume and a lower DTI-ALPS index than HCs adjusting for age and intracranial volume. Higher CP volume was associated with lower DTI-ALPS index, and longer disease duration. CONCLUSION Our findings demonstrate aberrant glymphatic function in FM, and that dysfunction in the brain glymphatic system may play a role in the neural mechanisms underlying FM.
Collapse
Affiliation(s)
- Ye Tu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiong
- Department of Radiology, PLA Central Theater General Hospital, Wuhan, China.
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
43
|
Cacciaguerra L, Rocca MA, Filippi M. Understanding the Pathophysiology and Magnetic Resonance Imaging of Multiple Sclerosis and Neuromyelitis Optica Spectrum Disorders. Korean J Radiol 2023; 24:1260-1283. [PMID: 38016685 PMCID: PMC10700997 DOI: 10.3348/kjr.2023.0360] [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: 04/26/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 11/30/2023] Open
Abstract
Magnetic resonance imaging (MRI) has been extensively applied in the study of multiple sclerosis (MS), substantially contributing to diagnosis, differential diagnosis, and disease monitoring. MRI studies have significantly contributed to the understanding of MS through the characterization of typical radiological features and their clinical or prognostic implications using conventional MRI pulse sequences and further with the application of advanced imaging techniques sensitive to microstructural damage. Interpretation of results has often been validated by MRI-pathology studies. However, the application of MRI techniques in the study of neuromyelitis optica spectrum disorders (NMOSD) remains an emerging field, and MRI studies have focused on radiological correlates of NMOSD and its pathophysiology to aid in diagnosis, improve monitoring, and identify relevant prognostic factors. In this review, we discuss the main contributions of MRI to the understanding of MS and NMOSD, focusing on the most novel discoveries to clarify differences in the pathophysiology of focal inflammation initiation and perpetuation, involvement of normal-appearing tissue, potential entry routes of pathogenic elements into the CNS, and existence of primary or secondary mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Laura Cacciaguerra
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milano, Italy.
| |
Collapse
|
44
|
Jeong SH, Jeong HJ, Sunwoo MK, Ahn SS, Lee SK, Lee PH, Kim YJ, Sohn YH, Park CJ, Chung SJ. Association between choroid plexus volume and cognition in Parkinson disease. Eur J Neurol 2023; 30:3114-3123. [PMID: 37498202 DOI: 10.1111/ene.15999] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/05/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND AND PURPOSE The choroid plexus (CP) clears harmful metabolites from the central nervous system as part of the glymphatic system. We investigated the association of CP volume (CPV) with baseline and longitudinal cognitive decline in patients with Parkinson disease (PD). METHODS We retrospectively reviewed the medical records of 240 patients with newly diagnosed PD who had undergone detailed neuropsychological tests and high-resolution T1-weighted structural magnetic resonance imaging during the initial assessment. The CPV of each patient was automatically segmented, and the intracranial volume ratio was used in subsequent analyses. The relationship between CPV and baseline composite scores of each cognitive domain was assessed using multivariate linear regression analyses. A Cox proportional hazards model was used to compare the risk of dementia conversion with CPV. RESULTS CPV negatively correlated with composite scores of the frontal/executive function domain (β = -0.375, p = 0.002) after adjusting for age, sex, years of education, and parkinsonian symptom duration. The Cox regression model revealed that a larger CPV was associated with a higher risk of dementia conversion (hazard ratio [HR] = 1.509, p = 0.038), which was no longer significant after adjusting for the composite scores of the frontal/executive function domain. A mediation analysis demonstrated that the effect of CPV on the risk of dementia conversion was completely mediated by frontal/executive function (direct effect: HR = 1.203, p = 0.396; indirect effect: HR = 1.400, p = 0.015). CONCLUSIONS Baseline CPV is associated with baseline frontal/executive function, which subsequently influences dementia conversion risk in patients with PD.
Collapse
Affiliation(s)
- Seong Ho Jeong
- Department of Neurology, Inje University Sanggye Paik Hospital, Seoul, Korea
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun-Jae Jeong
- Research Institute of Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Mun Kyung Sunwoo
- Department of Neurology, Bundang Jesaeng General Hospital, Seongnam-si, Korea
| | - Sung Soo Ahn
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science and Centre for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science and Centre for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Yun Joong Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea
- YONSEI BEYOND LAB, Yongin, Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Chae Jung Park
- Department of Radiology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea
| | - Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea
- YONSEI BEYOND LAB, Yongin, Korea
| |
Collapse
|
45
|
Jankowska A, Chwojnicki K, Grzywińska M, Trzonkowski P, Szurowska E. Choroid Plexus Volume Change-A Candidate for a New Radiological Marker of MS Progression. Diagnostics (Basel) 2023; 13:2668. [PMID: 37627928 PMCID: PMC10453931 DOI: 10.3390/diagnostics13162668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Multiple sclerosis (MS) is an auto-immune, chronic, neuroinflammatory, demyelinating disease that affects mainly young patients. This progressive inflammatory process causes the chronic loss of brain tissue and results in a deterioration in quality of life. To monitor neuroinflammatory process activity and predict the further development of disease, it is necessary to find a suitable biomarker that could easily be used. In this research, we verify the usability of choroid plexus (CP) volume, a new MS biomarker, in the monitoring of the progression of multiple sclerosis disease. (2) Methods: A single-center, prospective study with three groups of patients was conducted based on the following groups: MS patients who received experimental cellular therapy (Treg), treatment-naïve MS patients and healthy controls. (3) Results: This study concludes that there is a correlation between the CPV/TIV (choroid plexus/total intracranial volume) ratio and the progress of multiple sclerosis disease-patients with MS (MS + Treg) had larger volumes of choroid plexuses. CPV/TIV ratios in MS groups were constantly and significantly growing. In the Treg group, patients with relapses had larger plexuses in comparison to the group with no relapses of MS. A similar correlation was observed for the GD+ group (patients with postcontrast enhancing plaques) compared against the non-GD group (patients without postcontrast enhancing plaques). (4) Conclusion: Choroid plexus volume, due to its immunological function, correlates with the inflammatory process in the central nervous system. We consider it to become a valuable radiological biomarker of MS activity.
Collapse
Affiliation(s)
- Anna Jankowska
- 2nd Department of Radiology, Medical University of Gdańsk, Smoluchowskiego 17, 80-214 Gdańsk, Poland;
| | - Kamil Chwojnicki
- Department of Anesthesiology and Intensive Care, Medical University of Gdańsk, Debinki 7, 80-210 Gdańsk, Poland;
| | - Małgorzata Grzywińska
- Neuroinformatics and Artificial Intelligence Lab, Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdańsk, Debinki 7, 80-210 Gdańsk, Poland;
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Debinki 7, 80-210 Gdańsk, Poland;
| | - Edyta Szurowska
- 2nd Department of Radiology, Medical University of Gdańsk, Smoluchowskiego 17, 80-214 Gdańsk, Poland;
| |
Collapse
|
46
|
Williams MR, Macdonald CM, Turkheimer FE. Histological examination of choroid plexus epithelia changes in schizophrenia. Brain Behav Immun 2023; 111:292-297. [PMID: 37150267 DOI: 10.1016/j.bbi.2023.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/14/2023] [Accepted: 04/30/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND The choroid plexus (CP) produces and secretes most of the cerebrospinal fluid (CSF) of the central nervous system. The CP is suggested to be regulated by descending neurons and by circulating factors and is involved in the interaction between central and peripheral inflammation. Quantitative imaging has demonstrated volumetric CP changes in psychosis, schizophrenia and depression. This study histologically examines CP epithelial cell morphology in these illnesses to identify the biological source of such volumetric changes. METHODS Formalin-fixed paraffin-embedded (FFPE) blocks were obtained bilaterally from the lateral ventricles of 13 cases of sex- and age-matched brains from each of schizophrenia (SZ) with psychosis, major depressive disorder (MDD) and matched controls (NPD). FFPE blocks were sectioned at 7 μm and routinely stained for H&E. Morphological analysis of 180 CP epithelia/case was conducted blindly on digital images collected at x600 magnification. Calcification was assessed in all CP regions manually. RESULTS Analysis with a General Linear Model demonstrated a significant effect of diagnosis on somal width (p = 0.006, R2 = 0.33 R2(adj) = 0.25) demonstrating increased somal width in SZ without psychotic medication versus controls (p = 0.032), but not in medicated SZ cases. No effects were observed in calcification. DISCUSSION The epithelial cells that were examined were attached to the CP fibrous surface, so width expansion describes the primary methods for these cells to expand with adherence to this surface in SZ. The interaction of antipsychotic medication and diagnosis demonstrates that this is an illness-specific change mediated through the DA-system with likely neuronal origin. CP alterations were not found in MDD where they are instead generally associated with heightened allostatic load that was unknown in this cohort.
Collapse
Affiliation(s)
- M R Williams
- Segmentum Analysis, St John's Innovation Park, Cambridge Science Park, UK
| | | | - F E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
47
|
Butler T, Wang XH, Chiang GC, Li Y, Zhou L, Xi K, Wickramasuriya N, Tanzi E, Spector E, Ozsahin I, Mao X, Razlighi QR, Fung EK, Dyke JP, Maloney T, Gupta A, Raj A, Shungu DC, Mozley PD, Rusinek H, Glodzik L. Choroid Plexus Calcification Correlates with Cortical Microglial Activation in Humans: A Multimodal PET, CT, MRI Study. AJNR Am J Neuroradiol 2023; 44:776-782. [PMID: 37321857 PMCID: PMC10337614 DOI: 10.3174/ajnr.a7903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/04/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND PURPOSE The choroid plexus (CP) within the brain ventricles is well-known to produce cerebrospinal fluid (CSF). Recently, the CP has been recognized as critical in modulating inflammation. MRI-measured CP enlargement has been reported in neuroinflammatory disorders like MS as well as with aging and neurodegeneration. The basis of MRI-measured CP enlargement is unknown. On the basis of tissue studies demonstrating CP calcification as a common pathology associated with aging and disease, we hypothesized that previously unmeasured CP calcification contributes to MRI-measured CP volume and may be more specifically associated with neuroinflammation. MATERIALS AND METHODS We analyzed 60 subjects (43 healthy controls and 17 subjects with Parkinson's disease) who underwent PET/CT using 11C-PK11195, a radiotracer sensitive to the translocator protein expressed by activated microglia. Cortical inflammation was quantified as nondisplaceable binding potential. Choroid plexus calcium was measured via manual tracing on low-dose CT acquired with PET and automatically using a new CT/MRI method. Linear regression assessed the contribution of choroid plexus calcium, age, diagnosis, sex, overall volume of the choroid plexus, and ventricle volume to cortical inflammation. RESULTS Fully automated choroid plexus calcium quantification was accurate (intraclass correlation coefficient with manual tracing = .98). Subject age and choroid plexus calcium were the only significant predictors of neuroinflammation. CONCLUSIONS Choroid plexus calcification can be accurately and automatically quantified using low-dose CT and MRI. Choroid plexus calcification-but not choroid plexus volume-predicted cortical inflammation. Previously unmeasured choroid plexus calcium may explain recent reports of choroid plexus enlargement in human inflammatory and other diseases. Choroid plexus calcification may be a specific and relatively easily acquired biomarker for neuroinflammation and choroid plexus pathology in humans.
Collapse
Affiliation(s)
- T Butler
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - X H Wang
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - G C Chiang
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - Y Li
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - L Zhou
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - K Xi
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - N Wickramasuriya
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - E Tanzi
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - E Spector
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - I Ozsahin
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - X Mao
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - Q R Razlighi
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - E K Fung
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - J P Dyke
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - T Maloney
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - A Gupta
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - A Raj
- Department of Radiology (A.R.), University of California, San Francisco, San Francisco, California
| | - D C Shungu
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - P D Mozley
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - H Rusinek
- Department of Radiology (H.R.), New York University, New York, New York
| | - L Glodzik
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| |
Collapse
|
48
|
Turkheimer FE, Veronese M, Mondelli V, Cash D, Pariante CM. Sickness behaviour and depression: An updated model of peripheral-central immunity interactions. Brain Behav Immun 2023; 111:202-210. [PMID: 37076054 DOI: 10.1016/j.bbi.2023.03.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/15/2023] [Accepted: 03/26/2023] [Indexed: 04/21/2023] Open
Abstract
Current research into mood disorders indicates that circulating immune mediators participating in the pathophysiology of chronic somatic disorders have potent influences on brain function. This paradigm has brought to the fore the use of anti-inflammatory therapies as adjunctive to standard antidepressant therapy to improve treatment efficacy, particularly in subjects that do not respond to standard medication. Such new practice requires biomarkers to tailor these new therapies to those most likely to benefit but also validated mechanisms of action describing the interaction between peripheral immunity and brain function to optimize target intervention. These mechanisms are generally studied in preclinical models that try to recapitulate the human disease, MDD, through peripherally induced sickness behaviour. In this proposal paper, after an appraisal of the data in rodent models and their adherence to the data in clinical cohorts, we put forward a modified model of periphery-brain interactions that goes beyond the currently established view of microglia cells as the drivers of depression. Instead, we suggest that, for most patients with mild levels of peripheral inflammation, brain barriers are the primary actors in the pathophysiology of the disease and in treatment resistance. We then highlight data gaps in this proposal and suggest novel lines of research.
Collapse
Affiliation(s)
- Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Department of Information Engineering, University of Padova, Padova, Italy
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Diana Cash
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
49
|
Ricigliano VAG, Stankoff B. Choroid plexuses at the interface of peripheral immunity and tissue repair in multiple sclerosis. Curr Opin Neurol 2023; 36:214-221. [PMID: 37078651 DOI: 10.1097/wco.0000000000001160] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
PURPOSE OF REVIEW Choroid plexuses (ChPs) are key actors of the blood-to-cerebrospinal-fluid barrier and serve as brain immune checkpoint. The past years have seen a regain of interest about their potential involvement in the physiopathology of neuroinflammatory disorders like multiple sclerosis (MS). This article offers an overview of the recent findings on ChP alterations in MS, with a focus on the imaging tools able to detect these abnormalities and on their involvement in inflammation, tissue damage and repair. RECENT FINDINGS On MRI, ChPs are enlarged in people with MS (PwMS) versus healthy individuals. This size increase is an early event, already detected in presymptomatic and pediatric MS. Enlargement of ChPs is linked to local inflammatory infiltrates, and their dysfunction selectively impacts periventricular damage, larger ChPs predicting the expansion of chronic active lesions, smoldering inflammation and remyelination failure in tissues surrounding the ventricles. ChP volumetry may add value for the prediction of disease activity and disability worsening. SUMMARY ChP imaging metrics are emerging as possible biomarkers of neuroinflammation and repair failure in MS. Future works combining multimodal imaging techniques should provide a more refined characterization of ChP functional changes, their link with tissue damage, blood to cerebrospinal-fluid barrier dysfunction and fluid trafficking in MS.
Collapse
Affiliation(s)
- Vito A G Ricigliano
- Sorbonne Université, Paris Brain Institute, ICM, CNRS, Inserm
- Neurology Department, Pitié-Salpêtrière Hospital
| | - Bruno Stankoff
- Sorbonne Université, Paris Brain Institute, ICM, CNRS, Inserm
- Neurology Department, St Antoine Hospital, APHP-Sorbonne, Paris, France
| |
Collapse
|
50
|
Brasanac J, Chien C. A review on multiple sclerosis prognostic findings from imaging, inflammation, and mental health studies. Front Hum Neurosci 2023; 17:1151531. [PMID: 37250694 PMCID: PMC10213782 DOI: 10.3389/fnhum.2023.1151531] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
Magnetic resonance imaging (MRI) of the brain is commonly used to detect where chronic and active lesions are in multiple sclerosis (MS). MRI is also extensively used as a tool to calculate and extrapolate brain health by way of volumetric analysis or advanced imaging techniques. In MS patients, psychiatric symptoms are common comorbidities, with depression being the main one. Even though these symptoms are a major determinant of quality of life in MS, they are often overlooked and undertreated. There has been evidence of bidirectional interactions between the course of MS and comorbid psychiatric symptoms. In order to mitigate disability progression in MS, treating psychiatric comorbidities should be investigated and optimized. New research for the prediction of disease states or phenotypes of disability have advanced, primarily due to new technologies and a better understanding of the aging brain.
Collapse
Affiliation(s)
- Jelena Brasanac
- Charité – Universitätsmedizin Berlin, Klinik für Psychiatrie und Psychotherapie, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Medizinische Klinik m.S. Psychosomatik, Berlin, Germany
| | - Claudia Chien
- Charité – Universitätsmedizin Berlin, Klinik für Psychiatrie und Psychotherapie, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Medizinische Klinik m.S. Psychosomatik, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Neuroscience Clinical Research Center, Berlin, Germany
| |
Collapse
|