1
|
Krsek A, Ostojic L, Zivalj D, Baticic L. Navigating the Neuroimmunomodulation Frontier: Pioneering Approaches and Promising Horizons-A Comprehensive Review. Int J Mol Sci 2024; 25:9695. [PMID: 39273641 PMCID: PMC11396210 DOI: 10.3390/ijms25179695] [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: 07/31/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
The research in neuroimmunomodulation aims to shed light on the complex relationships that exist between the immune and neurological systems and how they affect the human body. This multidisciplinary field focuses on the way immune responses are influenced by brain activity and how neural function is impacted by immunological signaling. This provides important insights into a range of medical disorders. Targeting both brain and immunological pathways, neuroimmunomodulatory approaches are used in clinical pain management to address chronic pain. Pharmacological therapies aim to modulate neuroimmune interactions and reduce inflammation. Furthermore, bioelectronic techniques like vagus nerve stimulation offer non-invasive control of these systems, while neuromodulation techniques like transcranial magnetic stimulation modify immunological and neuronal responses to reduce pain. Within the context of aging, neuroimmunomodulation analyzes the ways in which immunological and neurological alterations brought on by aging contribute to cognitive decline and neurodegenerative illnesses. Restoring neuroimmune homeostasis through strategies shows promise in reducing age-related cognitive decline. Research into mood disorders focuses on how immunological dysregulation relates to illnesses including anxiety and depression. Immune system fluctuations are increasingly recognized for their impact on brain function, leading to novel treatments that target these interactions. This review emphasizes how interdisciplinary cooperation and continuous research are necessary to better understand the complex relationship between the neurological and immune systems.
Collapse
Affiliation(s)
- Antea Krsek
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Leona Ostojic
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Dorotea Zivalj
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
2
|
Mariottini A, Stack EH, Nair G, Nozzoli C, Wu T, Marchi L, Boncompagni R, Repice AM, Fainardi E, Pasquale FD, Carlesi E, Saccardi R, Jacobson S, Massacesi L. Spinal cord size as promising biomarker of disability outcomes after hematopoietic stem cell transplantation in multiple sclerosis. Mult Scler Relat Disord 2024; 88:105745. [PMID: 38996712 DOI: 10.1016/j.msard.2024.105745] [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: 01/30/2024] [Revised: 05/20/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Biomarkers predictive of disability outcomes in individual multiple sclerosis (MS) patients undergoing autologous haematopoietic stem cell transplantation (AHSCT) are currently lacking. As correlations between spinal cord atrophy and clinical disability in MS were previously described, in this study spinal cord size was investigated in MS patients treated with AHSCT, exploring whether baseline spinal cord volume may predict disability progression after AHSCT. METHODS relapsing-remitting (RR-) and secondary-progressive (SP-) MS patients treated with AHSCT (BEAM/ATG regimen) at a single academic centre in Florence, who performed at least two standardized brain magnetic resonance imaging (MRIs) scans (acquired between one-year pre-AHSCT to 5 years after AHSCT) were included. Cervical spinal cord atrophy was estimated as upper cervical spinal cord cross-sectional area (SCCSA). Brain volume loss (BVL) was analysed at the same timepoints. RESULTS Eleven (8 RR-; 3 SP-) MS patients were included. Over a median follow-up of 66 (range 37 - 100) months, no relapses nor brain MRI activity were observed; disability progressed in 2 cases (both SP-MS). Baseline SCCSA was associated with EDSS change between pre- and one-year post-AHSCT. Compared to patients who stabilized, patients who progressed after AHSCT tended to have lower SCCSA at C4 level at baseline and year 1 after AHSCT. Longitudinal changes in SCCSA or BVL did not correlate with EDSS change. CONCLUSIONS Baseline pre-AHSCT SCCSA, but not its longitudinal changes nor BVL, predicted EDSS change within the two years following AHSCT. SCCSA may represent a biomarker of treatment response and a promising screening tool for assessing patient eligibility for high-impact treatments such as AHSCT.
Collapse
Affiliation(s)
- Alice Mariottini
- Department of Neurofarba, University of Florence, Florence, Italy; Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Centre, Careggi University Hospital, Florence, Italy
| | - Emily H Stack
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892 USA
| | - Govind Nair
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892 USA
| | - Chiara Nozzoli
- Cell Therapy and Transfusion Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Tianxia Wu
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892 USA
| | - Leonardo Marchi
- Department of Neurofarba, University of Florence, Florence, Italy
| | - Riccardo Boncompagni
- Cell Therapy and Transfusion Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Anna Maria Repice
- Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Centre, Careggi University Hospital, Florence, Italy
| | - Enrico Fainardi
- Neuroradiology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy; Neuroradiology Unit, Department of Radiology, Careggi University Hospital, Florence, Italy
| | - Francesca Di Pasquale
- Diagnostic Imaging Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Edoardo Carlesi
- Neuroradiology Unit, Department of Radiology, Careggi University Hospital, Florence, Italy
| | - Riccardo Saccardi
- Cell Therapy and Transfusion Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Steven Jacobson
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892 USA
| | - Luca Massacesi
- Department of Neurofarba, University of Florence, Florence, Italy; Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Centre, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
3
|
Mascali D, Villani A, Chiarelli AM, Biondetti E, Lipp I, Digiovanni A, Pozzilli V, Caporale AS, Rispoli MG, Ajdinaj P, D'Apolito M, Grasso E, Sensi SL, Murphy K, Tomassini V, Wise RG. Pathophysiology of multiple sclerosis damage and repair: Linking cerebral hypoperfusion to the development of irreversible tissue loss in multiple sclerosis using magnetic resonance imaging. Eur J Neurol 2023; 30:2348-2356. [PMID: 37154298 PMCID: PMC7615142 DOI: 10.1111/ene.15827] [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: 12/23/2022] [Revised: 03/10/2023] [Accepted: 05/01/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND AND PURPOSE Reduced cerebral perfusion has been observed in multiple sclerosis (MS) and may contribute to tissue loss both acutely and chronically. Here, we test the hypothesis that hypoperfusion occurs in MS and relates to the presence of irreversible tissue damage. METHODS In 91 patients with relapsing MS and 26 healthy controls (HC), gray matter (GM) cerebral blood flow (CBF) was assessed using pulsed arterial spin labeling. GM volume, T1 hypointense and T2 hyperintense lesion volumes (T1LV and T2LV, respectively), and the proportion of T2-hyperintense lesion volume that appears hypointense on T1-weighted magnetic resonance imaging (T1LV/T2LV) were quantified. GM CBF and GM volume were evaluated globally, as well as regionally, using an atlas-based approach. RESULTS Global GM CBF was lower in patients (56.9 ± 12.3 mL/100 g/min) than in HC (67.7 ± 10.0 mL/100 g/min; p < 0.001), a difference that was widespread across brain regions. Although total GM volume was comparable between groups, significant reductions were observed in a subset of subcortical structures. GM CBF negatively correlated with T1LV (r = -0.43, p = 0.0002) and T1LV/T2LV (r = -0.37, p = 0.0004), but not with T2LV. CONCLUSIONS GM hypoperfusion occurs in MS and is associated with irreversible white matter damage, thus suggesting that cerebral hypoperfusion may actively contribute and possibly precede neurodegeneration by hampering tissue repair abilities in MS.
Collapse
Affiliation(s)
- Daniele Mascali
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical TechnologiesG. d'Annunzio University of Chieti‐PescaraChietiItaly
| | - Alessandro Villani
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical TechnologiesG. d'Annunzio University of Chieti‐PescaraChietiItaly
| | - Antonio M. Chiarelli
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical TechnologiesG. d'Annunzio University of Chieti‐PescaraChietiItaly
| | - Emma Biondetti
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical TechnologiesG. d'Annunzio University of Chieti‐PescaraChietiItaly
| | - Ilona Lipp
- Department of NeurophysicsMax Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
- Cardiff University Brain Research Imaging Centre, School of PsychologyCardiff UniversityCardiffUK
| | - Anna Digiovanni
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- MS Centre, Department of Clinical NeurologySS. Annunziata University HospitalChietiItaly
| | - Valeria Pozzilli
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- MS Centre, Department of Clinical NeurologySS. Annunziata University HospitalChietiItaly
| | - Alessandra S. Caporale
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical TechnologiesG. d'Annunzio University of Chieti‐PescaraChietiItaly
| | - Marianna G. Rispoli
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- MS Centre, Department of Clinical NeurologySS. Annunziata University HospitalChietiItaly
| | - Paola Ajdinaj
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- MS Centre, Department of Clinical NeurologySS. Annunziata University HospitalChietiItaly
| | - Maria D'Apolito
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- MS Centre, Department of Clinical NeurologySS. Annunziata University HospitalChietiItaly
| | - Eleonora Grasso
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Department of PaediatricsSS. Annunziata University HospitalChietiItaly
| | - Stefano L. Sensi
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical TechnologiesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Behavioral Neurology and Molecular Neurology Units, Centre for Advanced Studies and TechnologyG. d'Annunzio University of Chieti‐PescaraChietiItaly
| | - Kevin Murphy
- Cardiff University Brain Research Imaging Centre, School of Physics and AstronomyCardiff UniversityCardiffUK
| | - Valentina Tomassini
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical TechnologiesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Cardiff University Brain Research Imaging Centre, School of PsychologyCardiff UniversityCardiffUK
- MS Centre, Department of Clinical NeurologySS. Annunziata University HospitalChietiItaly
| | - Richard G. Wise
- Department of Neurosciences, Imaging, and Clinical SciencesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical TechnologiesG. d'Annunzio University of Chieti‐PescaraChietiItaly
- Cardiff University Brain Research Imaging Centre, School of PsychologyCardiff UniversityCardiffUK
| |
Collapse
|
4
|
Wang Y, Zhang Z, Li B, He B, Li L, Nice EC, Zhang W, Xu J. New Insights into the Gut Microbiota in Neurodegenerative Diseases from the Perspective of Redox Homeostasis. Antioxidants (Basel) 2022; 11:2287. [PMID: 36421473 PMCID: PMC9687622 DOI: 10.3390/antiox11112287] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 08/27/2023] Open
Abstract
An imbalance between oxidants and antioxidants in the body can lead to oxidative stress, which is one of the major causes of neurodegenerative diseases. The gut microbiota contains trillions of beneficial bacteria that play an important role in maintaining redox homeostasis. In the last decade, the microbiota-gut-brain axis has emerged as a new field that has revolutionized the study of the pathology, diagnosis, and treatment of neurodegenerative diseases. Indeed, a growing number of studies have found that communication between the brain and the gut microbiota can be accomplished through the endocrine, immune, and nervous systems. Importantly, dysregulation of the gut microbiota has been strongly associated with the development of oxidative stress-mediated neurodegenerative diseases. Therefore, a deeper understanding of the relationship between the gut microbiota and redox homeostasis will help explain the pathogenesis of neurodegenerative diseases from a new perspective and provide a theoretical basis for proposing new therapeutic strategies for neurodegenerative diseases. In this review, we will describe the role of oxidative stress and the gut microbiota in neurodegenerative diseases and the underlying mechanisms by which the gut microbiota affects redox homeostasis in the brain, leading to neurodegenerative diseases. In addition, we will discuss the potential applications of maintaining redox homeostasis by modulating the gut microbiota to treat neurodegenerative diseases, which could open the door for new therapeutic approaches to combat neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu Wang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhe Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bowen Li
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bo He
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Wei Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, China
| | - Jia Xu
- School of Medicine, Ningbo University, Ningbo 315211, China
| |
Collapse
|
5
|
Shaghaghi M, Cai K. Toward In Vivo MRI of the Tissue Proton Exchange Rate in Humans. BIOSENSORS 2022; 12:bios12100815. [PMID: 36290953 PMCID: PMC9599426 DOI: 10.3390/bios12100815] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/19/2022] [Accepted: 09/29/2022] [Indexed: 05/28/2023]
Abstract
Quantification of proton exchange rate (kex) is a challenge in MR studies. Current techniques either have low resolutions or are dependent on the estimation of parameters that are not measurable. The Omega plot method, on the other hand, provides a direct way for determining kex independent of the agent concentration. However, it cannot be used for in vivo studies without some modification due to the contributions from the water signal. In vivo tissue proton exchange rate (kex) MRI, based on the direct saturation (DS) removed Omega plot, quantifies the weighted average of kex of the endogenous tissue metabolites. This technique has been successfully employed for imaging the variation in the kex of ex vivo phantoms, as well as in vivo human brains in healthy subjects, and stroke or multiple sclerosis (MS) patients. In this paper, we present a brief review of the methods used for kex imaging with a focus on the development of in vivo kex MRI technique based on the DS-removed Omega plot. We then review the recent clinical studies utilizing this technique for better characterizing brain lesions. We also outline technical challenges for the presented technique and discuss its prospects for detecting tissue microenvironmental changes under oxidative stress.
Collapse
Affiliation(s)
- Mehran Shaghaghi
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Kejia Cai
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
6
|
Taloni A, Farrelly FA, Pontillo G, Petsas N, Giannì C, Ruggieri S, Petracca M, Brunetti A, Pozzilli C, Pantano P, Tommasin S. Evaluation of Disability Progression in Multiple Sclerosis via Magnetic-Resonance-Based Deep Learning Techniques. Int J Mol Sci 2022; 23:ijms231810651. [PMID: 36142563 PMCID: PMC9505100 DOI: 10.3390/ijms231810651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Short-term disability progression was predicted from a baseline evaluation in patients with multiple sclerosis (MS) using their three-dimensional T1-weighted (3DT1) magnetic resonance images (MRI). One-hundred-and-eighty-one subjects diagnosed with MS underwent 3T-MRI and were followed up for two to six years at two sites, with disability progression defined according to the expanded-disability-status-scale (EDSS) increment at the follow-up. The patients’ 3DT1 images were bias-corrected, brain-extracted, registered onto MNI space, and divided into slices along coronal, sagittal, and axial projections. Deep learning image classification models were applied on slices and devised as ResNet50 fine-tuned adaptations at first on a large independent dataset and secondly on the study sample. The final classifiers’ performance was evaluated via the area under the curve (AUC) of the false versus true positive diagram. Each model was also tested against its null model, obtained by reshuffling patients’ labels in the training set. Informative areas were found by intersecting slices corresponding to models fulfilling the disability progression prediction criteria. At follow-up, 34% of patients had disability progression. Five coronal and five sagittal slices had one classifier surviving the AUC evaluation and null test and predicted disability progression (AUC > 0.72 and AUC > 0.81, respectively). Likewise, fifteen combinations of classifiers and axial slices predicted disability progression in patients (AUC > 0.69). Informative areas were the frontal areas, mainly within the grey matter. Briefly, 3DT1 images may give hints on disability progression in MS patients, exploiting the information hidden in the MRI of specific areas of the brain.
Collapse
Affiliation(s)
- Alessandro Taloni
- Institute for Complex Systems, National Research Council (ISC-CNR), 00185 Rome, Italy
| | | | - Giuseppe Pontillo
- Department of Advanced Biomedical Sciences, Federico II University of Naples, 80131 Naples, Italy
- Department of Electrical Engineering and Information Technology, Federico II University of Naples, 80125 Naples, Italy
| | - Nikolaos Petsas
- Department of Radiology, IRCCS NEUROMED, 86077 Pozzilli, Italy
| | - Costanza Giannì
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Serena Ruggieri
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
- Neuroimmunology Unit, IRCSS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Maria Petracca
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University of Naples, 80131 Naples, Italy
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Carlo Pozzilli
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Patrizia Pantano
- Department of Radiology, IRCCS NEUROMED, 86077 Pozzilli, Italy
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Silvia Tommasin
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
7
|
Pitombeira MS, Koole M, Campanholo KR, Souza AM, Duran FLS, Solla DJF, Mendes MF, Pereira SLA, Rimkus CM, Busatto GF, Callegaro D, Buchpiguel CA, de Paula Faria D. Innate immune cells and myelin profile in multiple sclerosis: a multi-tracer PET/MR study. Eur J Nucl Med Mol Imaging 2022; 49:4551-4566. [PMID: 35838758 DOI: 10.1007/s00259-022-05899-2] [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: 12/02/2021] [Accepted: 06/30/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Neuropathological studies have demonstrated distinct profiles of microglia activation and myelin injury among different multiple sclerosis (MS) phenotypes and disability stages. PET imaging using specific tracers may uncover the in vivo molecular pathology and broaden the understanding of the disease heterogeneity. METHODS We used the 18-kDa translocator protein (TSPO) tracer (R)-[11C]PK11195 and [11C]PIB PET images acquired in a hybrid PET/MR 3 T system to characterize, respectively, the profile of innate immune cells and myelin content in 47 patients with MS compared to 18 healthy controls (HC). For the volume of interest (VOI)-based analysis of the dynamic data, (R)-[11C]PK11195 distribution volume (VT) was determined for each subject using a metabolite-corrected arterial plasma input function while [11C]PIB distribution volume ratio (DVR) was estimated using a reference region extracted by a supervised clustering algorithm. A voxel-based analysis was also performed using Statistical Parametric Mapping. Functional disability was evaluated by the Expanded Disability Status Scale (EDSS), Multiple Sclerosis Functional Composite (MSFC), and Symbol Digit Modality Test (SDMT). RESULTS In the VOI-based analysis, [11C]PIB DVR differed between patients and HC in the corpus callosum (P = 0.019) while no differences in (R)-[11C]PK11195 VT were observed in patients relative to HC. Furthermore, no correlations or associations were observed between both tracers within the VOI analyzed. In the voxel-based analysis, high (R)-[11C]PK11195 uptake was observed diffusively in the white matter (WM) when comparing the progressive phenotype and HC, and lower [11C]PIB uptake was observed in certain WM regions when comparing the relapsing-remitting phenotype and HC. None of the tracers were able to differentiate phenotypes at voxel or VOI level in our cohort. Linear regression models adjusted for age, sex, and phenotype demonstrated that higher EDSS was associated with an increased (R)-[11C]PK11195 VT and lower [11C]PIB DVR in corpus callosum (P = 0.001; P = 0.023), caudate (P = 0.015; P = 0.008), and total T2 lesion (P = 0.007; P = 0.012), while better cognitive scores in SDMT were associated with higher [11C]PIB DVR in the corpus callosum (P = 0.001), and lower (R)-[11C]PK11195 VT (P = 0.013). CONCLUSIONS Widespread innate immune cells profile and marked loss of myelin in T2 lesions and regions close to the ventricles may occur independently and are associated with disability, in both WM and GM structures.
Collapse
Affiliation(s)
- Milena Sales Pitombeira
- Department of Neurology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil.,Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Michel Koole
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Flanders, Belgium
| | - Kenia R Campanholo
- Department of Neurology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil.,Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Aline M Souza
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Fábio L S Duran
- Laboratory of Psychiatric Neuroimaging (LIM21), Department of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Davi J Fontoura Solla
- Department of Neurology, Division of Neurosurgery, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Maria F Mendes
- Department of Neurology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Carolina M Rimkus
- Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Geraldo Filho Busatto
- Laboratory of Psychiatric Neuroimaging (LIM21), Department of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Dagoberto Callegaro
- Department of Neurology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Carlos A Buchpiguel
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Daniele de Paula Faria
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
8
|
Soheili M, Alinaghipour A, Salami M. Good bacteria, oxidative stress and neurological disorders: Possible therapeutical considerations. Life Sci 2022; 301:120605. [DOI: 10.1016/j.lfs.2022.120605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022]
|
9
|
Swindell WR, Bojanowski K, Chaudhuri RK. Transcriptomic Analysis of Fumarate Compounds Identifies Unique Effects of Isosorbide Di-(Methyl Fumarate) on NRF2, NF-kappaB and IRF1 Pathway Genes. Pharmaceuticals (Basel) 2022; 15:ph15040461. [PMID: 35455458 PMCID: PMC9026097 DOI: 10.3390/ph15040461] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/09/2022] [Accepted: 04/10/2022] [Indexed: 11/16/2022] Open
Abstract
Dimethyl fumarate (DMF) has emerged as a first-line therapy for relapsing-remitting multiple sclerosis (RRMS). This treatment, however, has been limited by adverse effects, which has prompted development of novel derivatives with improved tolerability. We compared the effects of fumarates on gene expression in astrocytes. Our analysis included diroximel fumarate (DRF) and its metabolite monomethyl fumarate (MMF), along with a novel compound isosorbide di-(methyl fumarate) (IDMF). Treatment with IDMF resulted in the largest number of differentially expressed genes. The effects of DRF and MMF were consistent with NRF2 activation and NF-κB inhibition, respectively. IDMF responses, however, were concordant with both NRF2 activation and NF-κB inhibition, and we confirmed IDMF-mediated NF-κB inhibition using a reporter assay. IDMF also down-regulated IRF1 expression and IDMF-decreased gene promoters were enriched with IRF1 recognition sequences. Genes altered by each fumarate overlapped significantly with those near loci from MS genetic association studies, but IDMF had the strongest overall effect on MS-associated genes. These results show that next-generation fumarates, such as DRF and IDMF, have effects differing from those of the MMF metabolite. Our findings support a model in which IDMF attenuates oxidative stress via NRF2 activation, with suppression of NF-κB and IRF1 contributing to mitigation of inflammation and pyroptosis.
Collapse
Affiliation(s)
- William R. Swindell
- Department of Internal Medicine, The Jewish Hospital, Cincinnati, OH 45236, USA
- Correspondence:
| | - Krzysztof Bojanowski
- Sunny BioDiscovery Inc., Santa Paula, CA 93060, USA;
- Symbionyx Pharmaceuticals Inc., Boonton, NJ 07005, USA;
| | - Ratan K. Chaudhuri
- Symbionyx Pharmaceuticals Inc., Boonton, NJ 07005, USA;
- Sytheon Ltd., Boonton, NJ 07005, USA
| |
Collapse
|
10
|
Tommasin S, Iakovleva V, Rocca MA, Giannì C, Tedeschi G, De Stefano N, Pozzilli C, Filippi M, Pantano P. Relation of sensorimotor and cognitive cerebellum functional connectivity with brain structural damage in patients with multiple sclerosis and no disability. Eur J Neurol 2022; 29:2036-2046. [PMID: 35298059 PMCID: PMC9323479 DOI: 10.1111/ene.15329] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/11/2022] [Accepted: 02/27/2022] [Indexed: 12/01/2022]
Abstract
Background and purpose To investigate the relationship between the functional connectivity (FC) of the sensorimotor and cognitive cerebellum and measures of structural damage in patients with multiple sclerosis (MS) and no physical disability. Methods We selected 144 relapsing–remitting MS patients with an Expanded Disability Status Scale score of ≤1.5 and 98 healthy controls from the Italian Neuroimaging Network Initiative database. From multimodal 3T magnetic resonance imaging (MRI), including functional MRI at rest, we calculated lesion load, cortical thickness, and white matter, cortical gray matter, and caudate, putamen, thalamic, and cerebellar volumes. Voxel‐wise FC of the sensorimotor and cognitive cerebellum was assessed with seed‐based analysis, and multiple regression analysis was used to evaluate the relationship between FC and structural damage. Results Whole brain, white matter, caudate, putamen, and thalamic volumes were reduced in patients compared to controls, whereas cortical gray matter was not significantly different in patients versus controls. Both the sensorimotor and cognitive cerebellum showed a widespread pattern of increased and decreased FC that were negatively associated with structural measures, indicating that the lower the FC, the greater the tissue loss. Lastly, among multiple structural measures, cortical gray matter and white matter volumes were the best predictors of cerebellar FC alterations. Conclusions Increased and decreased cerebellar FC with several brain areas coexist in MS patients with no disability. Our data suggest that white matter loss hampers FC, whereas, in the absence of atrophy, cortical volume represents the framework for FC to increase.
Collapse
Affiliation(s)
- Silvia Tommasin
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | | | - Maria Assunta Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | | | - Gioacchino Tedeschi
- Department of Advanced Medical and Surgical Sciences and MRI-Center "SUN-FISM", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Carlo Pozzilli
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, 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
| | - Patrizia Pantano
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy.,IRCCS NEUROMED, Pozzilli, Italy
| | | |
Collapse
|
11
|
Meijboom R, Wiseman SJ, York EN, Bastin ME, Valdés Hernández MDC, Thrippleton MJ, Mollison D, White N, Kampaite A, Ng Kee Kwong K, Rodriguez Gonzalez D, Job D, Weaver C, Kearns PKA, Connick P, Chandran S, Waldman AD. Rationale and design of the brain magnetic resonance imaging protocol for FutureMS: a longitudinal multi-centre study of newly diagnosed patients with relapsing-remitting multiple sclerosis in Scotland. Wellcome Open Res 2022; 7:94. [PMID: 36865371 PMCID: PMC9971644 DOI: 10.12688/wellcomeopenres.17731.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2022] [Indexed: 12/22/2022] Open
Abstract
Introduction: Multiple sclerosis (MS) is a chronic neuroinflammatory and neurodegenerative disease. MS prevalence varies geographically and is notably high in Scotland. Disease trajectory varies significantly between individuals and the causes for this are largely unclear. Biomarkers predictive of disease course are urgently needed to allow improved stratification for current disease modifying therapies and future targeted treatments aimed at neuroprotection and remyelination. Magnetic resonance imaging (MRI) can detect disease activity and underlying damage non-invasively in vivo at the micro and macrostructural level. FutureMS is a prospective Scottish longitudinal multi-centre cohort study, which focuses on deeply phenotyping patients with recently diagnosed relapsing-remitting MS (RRMS). Neuroimaging is a central component of the study and provides two main primary endpoints for disease activity and neurodegeneration. This paper provides an overview of MRI data acquisition, management and processing in FutureMS. FutureMS is registered with the Integrated Research Application System (IRAS, UK) under reference number 169955. Methods and analysis: MRI is performed at baseline (N=431) and 1-year follow-up, in Dundee, Glasgow and Edinburgh (3T Siemens) and in Aberdeen (3T Philips), and managed and processed in Edinburgh. The core structural MRI protocol comprises T1-weighted, T2-weighted, FLAIR and proton density images. Primary imaging outcome measures are new/enlarging white matter lesions (WML) and reduction in brain volume over one year. Secondary imaging outcome measures comprise WML volume as an additional quantitative structural MRI measure, rim lesions on susceptibility-weighted imaging, and microstructural MRI measures, including diffusion tensor imaging and neurite orientation dispersion and density imaging metrics, relaxometry, magnetisation transfer (MT) ratio, MT saturation and derived g-ratio measures. Conclusions: FutureMS aims to reduce uncertainty around disease course and allow for targeted treatment in RRMS by exploring the role of conventional and advanced MRI measures as biomarkers of disease severity and progression in a large population of RRMS patients in Scotland.
Collapse
Affiliation(s)
- Rozanna Meijboom
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Stewart J. Wiseman
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Elizabeth N. York
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Mark E. Bastin
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Maria del C. Valdés Hernández
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Michael J. Thrippleton
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Daisy Mollison
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Nicole White
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Agniete Kampaite
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Koy Ng Kee Kwong
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - David Rodriguez Gonzalez
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Dominic Job
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Christine Weaver
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
| | - Patrick K. A. Kearns
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
| | - Peter Connick
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
| | - Adam D. Waldman
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
12
|
The blood-brain barrier in aging and neurodegeneration. Mol Psychiatry 2022; 27:2659-2673. [PMID: 35361905 PMCID: PMC9156404 DOI: 10.1038/s41380-022-01511-z] [Citation(s) in RCA: 178] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/24/2022] [Accepted: 02/24/2022] [Indexed: 12/01/2022]
Abstract
The blood-brain barrier (BBB) is vital for maintaining brain homeostasis by enabling an exquisite control of exchange of compounds between the blood and the brain parenchyma. Moreover, the BBB prevents unwanted toxins and pathogens from entering the brain. This barrier, however, breaks down with age and further disruption is a hallmark of many age-related disorders. Several drugs have been explored, thus far, to protect or restore BBB function. With the recent connection between the BBB and gut microbiota, microbial-derived metabolites have been explored for their capabilities to protect and restore BBB physiology. This review, will focus on the vital components that make up the BBB, dissect levels of disruption of the barrier, and discuss current drugs and therapeutics that maintain barrier integrity and the recent discoveries of effects microbial-derived metabolites have on BBB physiology.
Collapse
|
13
|
Abstract
Neuropsychiatric sequalae to coronavirus disease 2019 (COVID-19) infection are beginning to emerge, like previous Spanish influenza and severe acute respiratory syndrome episodes. Streptococcal infection in paediatric patients causing obsessive compulsive disorder (PANDAS) is another recent example of an infection-based psychiatric disorder. Inflammation associated with neuropsychiatric disorders has been previously reported but there is no standard clinical management approach established. Part of the reason is that it is unclear what factors determine the specific neuronal vulnerability and the efficacy of anti-inflammatory treatment in neuroinflammation. The emerging COVID-19 data suggested that in the acute stage, widespread neuronal damage appears to be the result of abnormal and overactive immune responses and cytokine storm is associated with poor prognosis. It is still too early to know if there are long-term-specific neuronal or brain regional damages associated with COVID-19, resulting in distinct neuropsychiatric disorders. In several major psychiatric disorders where neuroinflammation is present, patients with abnormal inflammatory markers may also experience less than favourable response or treatment resistance when standard treatment is used alone. Evidence regarding the benefits of co-administered anti-inflammatory agents such as COX-2 inhibitor is encouraging in selected patients though may not benefit others. Disease-modifying therapies are increasingly being applied to neuropsychiatric diseases characterised by abnormal or hyperreactive immune responses. Adjunct anti-inflammatory treatment may benefit selected patients and is definitely an important component of clinical management in the presence of neuroinflammation.
Collapse
|
14
|
Altered sensorimotor integration in multiple sclerosis: A combined neurophysiological and functional MRI study. Clin Neurophysiol 2021; 132:2191-2198. [PMID: 34293529 DOI: 10.1016/j.clinph.2021.05.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 04/02/2021] [Accepted: 05/25/2021] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To explore whether abnormal thalamic resting-state functional connectivity (rsFC) contributes to altered sensorimotor integration and hand dexterity impairment in multiple sclerosis (MS). METHODS To evaluate sensorimotor integration, we recorded kinematic features of index finger abductions during somatosensory temporal discrimination threshold (STDT) testing in 36 patients with relapsing-remitting MS and 39 healthy controls (HC). Participants underwent a multimodal 3T structural and functional MRI protocol. RESULTS Patients had lower index finger abduction velocity during STDT testing compared to HC. Thalamic rsFC with the precentral and postcentral gyri, supplementary motor area (SMA), insula, and basal ganglia was higher in patients than HC. Intrathalamic rsFC and thalamic rsFC with caudate and insula bilaterally was lower in patients than HC. Finger movement velocity positively correlated with intrathalamic rsFC and negatively correlated with thalamic rsFC with the precentral and postcentral gyri, SMA, and putamen. CONCLUSIONS Abnormal thalamic rsFC is a possible substrate for altered sensorimotor integration in MS, with high intrathalamic rsFC facilitating finger movements and increased thalamic rsFC with the basal ganglia and sensorimotor cortex contributing to motor performance deterioration. SIGNIFICANCE The combined study of thalamic functional connectivity and upper limb sensorimotor integration may be useful in identifying patients who can benefit from early rehabilitation to prevent upper limb motor impairment.
Collapse
|
15
|
Shahrampour S, Heholt J, Wang A, Vedaei F, Mohamed FB, Alizadeh M, Wang Z, Zabrecky G, Wintering N, Bazzan AJ, Leist TP, Monti DA, Newberg AB. N-acetyl cysteine administration affects cerebral blood flow as measured by arterial spin labeling MRI in patients with multiple sclerosis. Heliyon 2021; 7:e07615. [PMID: 34377857 PMCID: PMC8327674 DOI: 10.1016/j.heliyon.2021.e07615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/23/2021] [Accepted: 07/14/2021] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND The purpose of this study was to explore if administration of N-acetyl-cysteine (NAC) in patients with multiple sclerosis (MS) resulted in altered cerebral blood flow (CBF) based on Arterial Spin Labeling (ASL) magnetic resonance imaging (MRI). METHODS Twenty-three patients with mild to moderate MS, (17 relapsing remitting and 6 primary progressive) were randomized to either NAC plus standard of care (N = 11), or standard of care only (N = 12). The experimental group received NAC intravenously (50 mg/kg) once per week and orally (500mg 2x/day) the other six days. Patients in both groups were evaluated initially and after 2 months (of receiving the NAC or waitlist control) with ASL MRI to measure CBF. Clinical symptom questionnaires were also completed at both time points. RESULTS The CBF data showed significant differences in several brain regions including the pons, midbrain, left temporal and frontal lobe, left thalamus, right middle frontal lobe and right temporal/hippocampus (p < 0.001) in the MS group after treatment with NAC, when compared to the control group. Self-reported scores related to cognition and attention were also significantly improved in the NAC group as compared to the control group. CONCLUSIONS The results of this study suggest that NAC administration alters resting CBF in MS patients, and this is associated with qualitative improvements in cognition and attention. Given these findings, large scale efficacy studies will be of value to determine the potential clinical impact of NAC over the course of illness in patients with MS, as well as the most effective dosages and differential effects across subpopulations.
Collapse
Affiliation(s)
- Shiva Shahrampour
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Justin Heholt
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew Wang
- Charles E. Schmidt College of Medicine, Marcus Institute of Integrative Health at FAU Medicine, Florida Atlantic University, Boca Raton, FL USA
| | - Faezeh Vedaei
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Feroze B. Mohamed
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mahdi Alizadeh
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ze Wang
- Department of Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - George Zabrecky
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nancy Wintering
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anthony J. Bazzan
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Thomas P. Leist
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Daniel A. Monti
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew B. Newberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
16
|
Tommasin S, Cocozza S, Taloni A, Giannì C, Petsas N, Pontillo G, Petracca M, Ruggieri S, De Giglio L, Pozzilli C, Brunetti A, Pantano P. Machine learning classifier to identify clinical and radiological features relevant to disability progression in multiple sclerosis. J Neurol 2021; 268:4834-4845. [PMID: 33970338 PMCID: PMC8563671 DOI: 10.1007/s00415-021-10605-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 01/22/2023]
Abstract
Objectives To evaluate the accuracy of a data-driven approach, such as machine learning classification, in predicting disability progression in MS. Methods We analyzed structural brain images of 163 subjects diagnosed with MS acquired at two different sites. Participants were followed up for 2–6 years, with disability progression defined according to the expanded disability status scale (EDSS) increment at follow-up. T2-weighted lesion load (T2LL), thalamic and cerebellar gray matter (GM) volumes, fractional anisotropy of the normal appearing white matter were calculated at baseline and included in supervised machine learning classifiers. Age, sex, phenotype, EDSS at baseline, therapy and time to follow-up period were also included. Classes were labeled as stable or progressed disability. Participants were randomly chosen from both sites to build a sample including 50% patients showing disability progression and 50% patients being stable. One-thousand machine learning classifiers were applied to the resulting sample, and after testing for overfitting, classifier confusion matrix, relative metrics and feature importance were evaluated. Results At follow-up, 36% of participants showed disability progression. The classifier with the highest resulting metrics had accuracy of 0.79, area under the true positive versus false positive rates curve of 0.81, sensitivity of 0.90 and specificity of 0.71. T2LL, thalamic volume, disability at baseline and administered therapy were identified as important features in predicting disability progression. Classifiers built on radiological features had higher accuracy than those built on clinical features. Conclusions Disability progression in MS may be predicted via machine learning classifiers, mostly evaluating neuroradiological features. Supplementary Information The online version contains supplementary material available at 10.1007/s00415-021-10605-7.
Collapse
Affiliation(s)
- Silvia Tommasin
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy.
| | - Sirio Cocozza
- Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Alessandro Taloni
- Institute for Complex Systems, Italian National Research Council, Rome, Italy
| | - Costanza Giannì
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | | | - Giuseppe Pontillo
- Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli Federico II, Naples, Italy.,Dipartimento di Ingegneria Elettrica e delle Tecnologie dell'Informazione, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Maria Petracca
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy.,Dipartimento di Neuroscienze, Scienze Riproduttive e Odontostomatologiche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Serena Ruggieri
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy.,Neuroimmunology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Laura De Giglio
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy.,Neurology Unit, Medicine Department, San Filippo Neri Hospital, Rome, Italy
| | - Carlo Pozzilli
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Arturo Brunetti
- Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Patrizia Pantano
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy.,Department of Radiology, IRCCS NEUROMED, Pozzilli, Italy
| |
Collapse
|
17
|
The Role of Extracellular Vesicles in Demyelination of the Central Nervous System. Int J Mol Sci 2020; 21:ijms21239111. [PMID: 33266211 PMCID: PMC7729475 DOI: 10.3390/ijms21239111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/21/2022] Open
Abstract
It is being increasingly demonstrated that extracellular vesicles (EVs) are deeply involved in the physiology of the central nervous system (CNS). Processes such as synaptic activity, neuron-glia communication, myelination and immune response are modulated by EVs. Likewise, these vesicles may participate in many pathological processes, both as triggers of disease or, on the contrary, as mechanisms of repair. EVs play relevant roles in neurodegenerative disorders such as Alzheimer’s or Parkinson’s diseases, in viral infections of the CNS and in demyelinating pathologies such as multiple sclerosis (MS). This review describes the involvement of these membrane vesicles in major demyelinating diseases, including MS, neuromyelitis optica, progressive multifocal leukoencephalopathy and demyelination associated to herpesviruses.
Collapse
|
18
|
CSF1R signaling is a regulator of pathogenesis in progressive MS. Cell Death Dis 2020; 11:904. [PMID: 33097690 PMCID: PMC7584629 DOI: 10.1038/s41419-020-03084-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
Microglia serve as the innate immune cells of the central nervous system (CNS) by providing continuous surveillance of the CNS microenvironment and initiating defense mechanisms to protect CNS tissue. Upon injury, microglia transition into an activated state altering their transcriptional profile, transforming their morphology, and producing pro-inflammatory cytokines. These activated microglia initially serve a beneficial role, but their continued activation drives neuroinflammation and neurodegeneration. Multiple sclerosis (MS) is a chronic, inflammatory, demyelinating disease of the CNS, and activated microglia and macrophages play a significant role in mediating disease pathophysiology and progression. Colony-stimulating factor-1 receptor (CSF1R) and its ligand CSF1 are elevated in CNS tissue derived from MS patients. We performed a large-scale RNA-sequencing experiment and identified CSF1R as a key node of disease progression in a mouse model of progressive MS. We hypothesized that modulating microglia and infiltrating macrophages through the inhibition of CSF1R will attenuate deleterious CNS inflammation and reduce subsequent demyelination and neurodegeneration. To test this hypothesis, we generated a novel potent and selective small-molecule CSF1R inhibitor (sCSF1Rinh) for preclinical testing. sCSF1Rinh blocked receptor phosphorylation and downstream signaling in both microglia and macrophages and altered cellular functions including proliferation, survival, and cytokine production. In vivo, CSF1R inhibition with sCSF1Rinh attenuated neuroinflammation and reduced microglial proliferation in a murine acute LPS model. Furthermore, the sCSF1Rinh attenuated a disease-associated microglial phenotype and blocked both axonal damage and neurological impairments in an experimental autoimmune encephalomyelitis (EAE) model of MS. While previous studies have focused on microglial depletion following CSF1R inhibition, our data clearly show that signaling downstream of this receptor can be beneficially modulated in the context of CNS injury. Together, these data suggest that CSF1R inhibition can reduce deleterious microglial proliferation and modulate microglial phenotypes during neuroinflammatory pathogenesis, particularly in progressive MS.
Collapse
|
19
|
Almutairi AD, Hassan HA, Suppiah S, Alomair OI, Alshoaibi A, Almutairi H, Mahmud R. Lesion load assessment among multiple sclerosis patient using DIR, FLAIR, and T2WI sequences. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2020. [DOI: 10.1186/s43055-020-00312-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Magnetic resonance imaging (MRI) is one of the diagnostic imaging modalities employing in lesion detection in neurological disorders such as multiple sclerosis (MS). Advances in MRI techniques such as double inversion recovery (DIR) made it more sensitive to distinguish lesions in the brain. To investigate the lesion load on different anatomical regions of the brain with MS using DIR, fluid attenuated inversion recovery (FLAIR) and T2-weighted imaging (T2WI) sequences. A total of 97 MS patients were included in our retrospective study, confirmed by neurologist. The patients were randomly selected from the major hospital in Saudi Arabia. All images were obtained using 3T Scanner (Siemens Skyra). The images from the DIR, FLAIR, and T2WI sequence were compared on axial planes with identical anatomic position and the number of lesions was assigned to their anatomical region.
Results
Comparing the lesion load measurement at various brain anatomical regions showed a significant difference among those three methods (p < 0.05).
Conclusion
DIR is a valuable MRI sequence for better delineation, greater contrast measurements and the increasing total number of MS lesions in MRI, compared with FLAIR, and T2WI and DIR revealed more intracortical lesions as well; therefore, in MS patients, it is recommended to add DIR sequence in daily routine imaging sequences.
Collapse
|
20
|
Ye H, Shaghaghi M, Chen Q, Zhang Y, Lutz SE, Chen W, Cai K. In Vivo Proton Exchange Rate (k ex ) MRI for the Characterization of Multiple Sclerosis Lesions in Patients. J Magn Reson Imaging 2020; 53:408-415. [PMID: 32975008 DOI: 10.1002/jmri.27363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Currently available radiological methods do not completely capture the diversity of multiple sclerosis (MS) lesion subtypes. This lack of information hampers the understanding of disease progression and potential treatment stratification. For example, inflammation persists in some lesions after gadolinium (Gd) enhancement resolves. Novel metabolic and molecular imaging methods may improve the current assessments of MS pathophysiology. PURPOSE To compare the in vivo proton exchange rate (kex ) MRI with Gd-enhanced MRI for characterizing MS lesions. STUDY TYPE Retrospective. SUBJECTS Sixteen consecutively diagnosed relapsing-remitting multiple sclerosis (RRMS) patients. FIELD STRENGTH/SEQUENCE 3.0T MRI with T2 -weighted imaging, postcontrast T1 -weighted imaging, and single-slice chemical exchange saturation transfer imaging. ASSESSMENT MS lesions in white matter were assessed for Gd enhancement and kex elevation compared to normal-appearing white matter (NAWM). STATISTICAL TESTS Student's t-test was used for analyzing the difference of kex values between lesions and NAWM, with statistical significance set at 0.05. RESULTS Of all 153 MS lesions, 78 (51%) lesions were Gd-enhancing and 75 (49%) were Gd-negative. Without exception, all 78 Gd-enhancing lesions showed significantly elevated kex values compared to NAWM (924 ± 130 s-1 vs. 735 ± 61 s-1 , P < 0.05). Of 75 Gd-negative lesions, 18 lesions (24%) showed no kex elevation (762 ± 29 s-1 vs. 755 ± 28 s-1 , P = 0.47) and 57 (76%) showed significant kex elevation (950 ± 124 s-1 vs. 759 ± 48 s-1 , P < 0.05) compared to NAWM. MS lesions with kex elevation appeared nodular (118, 87.4%), ring-like (15, 11.1%), or irregular-shaped (2, 1.5%). DATA CONCLUSION For Gd-enhancing lesions, kex MRI is highly consistent with Gd-enhanced images by showing 100% of elevated kex . For all Gd-negative lesions, the discrepancy on kex MRI may further differentiate active slowly expanding lesions or chronic inactive lesions, supporting kex as an imaging biomarker for tissue oxidative stress and inflammation. Level of Evidence 2 Technical Efficacy Stage 3 J. MAGN. RESON. IMAGING 2021;53:408-415.
Collapse
Affiliation(s)
- Haiqi Ye
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mehran Shaghaghi
- Department of Radiology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Qianlan Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sarah E Lutz
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Weiwei Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kejia Cai
- Department of Radiology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, USA.,Center for MR Research, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
21
|
Pasqua G, Tommasin S, Bharti K, Ruggieri S, Petsas N, Piervincenzi C, Pozzilli C, Pantano P. Resting-state functional connectivity of anterior and posterior cerebellar lobes is altered in multiple sclerosis. Mult Scler 2020; 27:539-548. [PMID: 32463319 DOI: 10.1177/1352458520922770] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Damage to the cerebellar sensorimotor and cognitive domains may underlie physical and cognitive disability. OBJECTIVE To investigate resting-state functional connectivity (FC) of sensorimotor and cognitive cerebellum, and clinical correlates in multiple sclerosis (MS). METHODS A total of 119 patients with MS and 42 healthy subjects underwent multimodal 3T-magnetic resonance imaging (MRI). Patients were evaluated using the Expanded Disability Status Scale and Multiple Sclerosis Functional Composite Scale. After parcellation of sensorimotor (lobules I-V + VIII) and cognitive cerebellum (lobules VI, VII, IX, X), we calculated cerebellar resting-state FC using a seed-based approach. RESULTS In patients with MS, the sensorimotor cerebellum showed increased FC mainly with cerebellar, thalamic, and cortical (frontal, parietal, temporal) areas and decreased FC with insular areas; the cognitive cerebellum showed increased FC mainly with thalamic and cortical (temporal-occipital) areas, and decreased FC with frontal-insular areas. Both sensorimotor and cognitive cerebellar FC negatively correlated with disability, and positively with cognitive scores. Cerebellar structural damage only partially influenced results. CONCLUSION The two neocerebellar circuits showed altered FC with subcortical and cortical areas. The association between increased sensorimotor and cognitive cerebellar FC and low levels of physical and cognitive disability suggests that altered FC might modulate the effects of cerebellar structural damage on clinical condition.
Collapse
Affiliation(s)
- Gabriele Pasqua
- Medicine and Health Science Department, University of Molise, Campobasso, Italy/Human Neuroscience Department, Sapienza University of Rome, Rome, Italy
| | - Silvia Tommasin
- Human Neuroscience Department, Sapienza University of Rome, Rome, Italy
| | - Komal Bharti
- Human Neuroscience Department, Sapienza University of Rome, Rome, Italy
| | - Serena Ruggieri
- Human Neuroscience Department, Sapienza University of Rome, Rome, Italy
| | | | | | - Carlo Pozzilli
- Human Neuroscience Department, Sapienza University of Rome, Rome, Italy/Multiple Sclerosis Centre, Azienda Ospedaliera Sant'Andrea, Rome, Italy
| | - Patrizia Pantano
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy/IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
22
|
Monti DA, Zabrecky G, Leist TP, Wintering N, Bazzan AJ, Zhan T, Newberg AB. N-acetyl Cysteine Administration Is Associated With Increased Cerebral Glucose Metabolism in Patients With Multiple Sclerosis: An Exploratory Study. Front Neurol 2020; 11:88. [PMID: 32117038 PMCID: PMC7033492 DOI: 10.3389/fneur.2020.00088] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/24/2020] [Indexed: 01/01/2023] Open
Abstract
Background: Multiple Sclerosis (MS) is an autoimmune disease marked by progressive neurocognitive injury. Treatment options affording neuroprotective effects remain largely experimental. The purpose of this proof of concept study was to explore the effects of N-acetyl-cysteine (NAC) on cerebral glucose metabolism (CMRGlu) and symptoms in patients with multiple sclerosis (MS). Methods: Twenty-four patients with MS were randomized to either NAC plus standard of care, or standard of care only (waitlist control). The experimental group received NAC intravenously once per week and orally the other 6 days. Patients in both groups were evaluated at baseline and after 2 months (of receiving the NAC or waitlist control period) with an integrated Position Emission Tomography (PET)/ Magnetic Resonance Imaging (MRI) scanner, using 18F Fluorodeoxyglucose (FDG) to measure cerebral glucose metabolism. Following imaging evaluation at 2 months, subjects initially attributed to the standard of care arm were eligible for treatment with NAC. Clinical and symptom questionnaires were also completed initially and after 2 months. Results: The FDG PET data showed significantly increased cerebral glucose metabolism in several brain regions including the caudate, inferior frontal gyrus, lateral temporal gyrus, and middle temporal gyrus (p < 0.05) in the MS group treated with NAC, as compared to the control group. Self-reported scores related to cognition and attention were also significantly improved in the NAC group as compared to the control group. Conclusions: The results of this study suggest that NAC positively affects cerebral glucose metabolism in MS patients, which is associated with qualitative, patient reported improvements in cognition and attention. Larger scale studies may help to determine the clinical impact of NAC on measures of functioning over the course of illness, as well as the most effective dosage and dosage regimen.
Collapse
Affiliation(s)
- Daniel A Monti
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - George Zabrecky
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Thomas P Leist
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Nancy Wintering
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Anthony J Bazzan
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Tingting Zhan
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, United States
| | - Andrew B Newberg
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States.,Division of Nuclear Medicine, Department of Radiology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
23
|
Wang S, Millward JM, Hanke-Vela L, Malla B, Pilch K, Gil-Infante A, Waiczies S, Mueller S, Boehm-Sturm P, Guo J, Sack I, Infante-Duarte C. MR Elastography-Based Assessment of Matrix Remodeling at Lesion Sites Associated With Clinical Severity in a Model of Multiple Sclerosis. Front Neurol 2020; 10:1382. [PMID: 31998225 PMCID: PMC6970413 DOI: 10.3389/fneur.2019.01382] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022] Open
Abstract
Magnetic resonance imaging (MRI) with gadolinium based contrast agents (GBCA) is routinely used in the clinic to visualize lesions in multiple sclerosis (MS). Although GBCA reveal endothelial permeability, they fail to expose other aspects of lesion formation such as the magnitude of inflammation or tissue changes occurring at sites of blood-brain barrier (BBB) disruption. Moreover, evidence pointing to potential side effects of GBCA has been increasing. Thus, there is an urgent need to develop GBCA-independent imaging tools to monitor pathology in MS. Using MR-elastography (MRE), we previously demonstrated in both MS and the animal model experimental autoimmune encephalomyelitis (EAE) that inflammation was associated with a reduction of brain stiffness. Now, using the relapsing-remitting EAE model, we show that the cerebellum—a region with predominant inflammation in this model—is especially prone to loss of stiffness. We also demonstrate that, contrary to GBCA-MRI, reduction of brain stiffness correlates with clinical disability and is associated with enhanced expression of the extracellular matrix protein fibronectin (FN). Further, we show that FN is largely expressed by activated astrocytes at acute lesions, and reflects the magnitude of tissue remodeling at sites of BBB breakdown. Therefore, MRE could emerge as a safe tool suitable to monitor disease activity in MS.
Collapse
Affiliation(s)
- Shuangqing Wang
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Berlin, Germany.,Department of Neurology, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen, China
| | - Jason M Millward
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Berlin, Germany.,Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Laura Hanke-Vela
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Berlin, Germany
| | - Bimala Malla
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Berlin, Germany
| | - Kjara Pilch
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Berlin, Germany
| | - Ana Gil-Infante
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Berlin, Germany
| | - Sonia Waiczies
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Susanne Mueller
- Department of Experimental Neurology and Center for Stroke Research, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Experimental Neurology and Center for Stroke Research, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jing Guo
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ingolf Sack
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carmen Infante-Duarte
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Berlin, Germany
| |
Collapse
|
24
|
Lagana MM, Pelizzari L, Baglio F. Relationship between MRI perfusion and clinical severity in multiple sclerosis. Neural Regen Res 2020; 15:646-652. [PMID: 31638086 PMCID: PMC6975150 DOI: 10.4103/1673-5374.266906] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Perfusion alterations within several brain regions have been shown in multiple sclerosis patients using different magnetic resonance imaging (MRI) techniques. Furthermore, MRI-derived brain perfusion metrics have been investigated in association with multiple sclerosis phenotypes, physical disability, and cognitive impairment. However, a review focused on these aspects is still missing. Our aim was to review all the studies investigating the relationship between perfusion MRI and clinical severity during the last fifteen years to understand the clinical relevance of these findings. Perfusion differences among phenotypes were observed both with 1.5T and 3T scanners, with progressive multiple sclerosis presenting with lower perfusion values than relapsing-remitting multiple sclerosis patients. However, only 3T scanners showed a statistically significant distinction. Controversial results about the association between MRI-derived perfusion metrics and physical disability scores were found. However, the majority of the studies showed that lower brain perfusion and longer transit time are associated with more severe physical disability and worse cognitive performances.
Collapse
|
25
|
Walczak P. Imaging Myeloperoxidase in Demyelinating Lesions: Biomarker with Clinical Value for Multiple Sclerosis or Merely a Tool for Animal Research? Radiology 2019; 293:166-167. [PMID: 31483203 DOI: 10.1148/radiol.2019191777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Piotr Walczak
- From the Division of MR Research, Associate Professor, The Russell H. Morgan Department of Radiology and Radiological Science, Institute for Cell Engineering, Johns Hopkins University School of Medicine, 733 N Broadway, Broadway Research Building Room 647, Baltimore, Md 21205
| |
Collapse
|
26
|
Nutma E, Willison H, Martino G, Amor S. Neuroimmunology - the past, present and future. Clin Exp Immunol 2019; 197:278-293. [PMID: 30768789 PMCID: PMC6693969 DOI: 10.1111/cei.13279] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
Neuroimmunology as a separate discipline has its roots in the fields of neurology, neuroscience and immunology. Early studies of the brain by Golgi and Cajal, the detailed clinical and neuropathology studies of Charcot and Thompson's seminal paper on graft acceptance in the central nervous system, kindled a now rapidly expanding research area, with the aim of understanding pathological mechanisms of inflammatory components of neurological disorders. While neuroimmunologists originally focused on classical neuroinflammatory disorders, such as multiple sclerosis and infections, there is strong evidence to suggest that the immune response contributes to genetic white matter disorders, epilepsy, neurodegenerative diseases, neuropsychiatric disorders, peripheral nervous system and neuro-oncological conditions, as well as ageing. Technological advances have greatly aided our knowledge of how the immune system influences the nervous system during development and ageing, and how such responses contribute to disease as well as regeneration and repair. Here, we highlight historical aspects and milestones in the field of neuroimmunology and discuss the paradigm shifts that have helped provide novel insights into disease mechanisms. We propose future perspectives including molecular biological studies and experimental models that may have the potential to push many areas of neuroimmunology. Such an understanding of neuroimmunology will open up new avenues for therapeutic approaches to manipulate neuroinflammation.
Collapse
Affiliation(s)
- E. Nutma
- Department of PathologyAmsterdam UMC, VUmc siteAmsterdamthe Netherlands
| | - H. Willison
- University of Glasgow, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesGlasgowUK
| | - G. Martino
- Neuroimmunology Unit, Division of NeuroscienceInstitute of Experimental Neurology (INSpe), San Raffaele Scientific Institute and Vita‐Salute San Raffaele UniversityMilanItaly
| | - S. Amor
- Department of PathologyAmsterdam UMC, VUmc siteAmsterdamthe Netherlands
- Centre for Neuroscience and TraumaThe Blizard Institute, Barts and The London School of Medicine and DentistryLondonUK
| |
Collapse
|
27
|
Wimmer I, Tietz S, Nishihara H, Deutsch U, Sallusto F, Gosselet F, Lyck R, Muller WA, Lassmann H, Engelhardt B. PECAM-1 Stabilizes Blood-Brain Barrier Integrity and Favors Paracellular T-Cell Diapedesis Across the Blood-Brain Barrier During Neuroinflammation. Front Immunol 2019; 10:711. [PMID: 31024547 PMCID: PMC6460670 DOI: 10.3389/fimmu.2019.00711] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 03/15/2019] [Indexed: 01/13/2023] Open
Abstract
Breakdown of the blood-brain barrier (BBB) and increased immune cell trafficking into the central nervous system (CNS) are hallmarks of the pathogenesis of multiple sclerosis (MS). Platelet endothelial cell adhesion molecule-1 (PECAM-1; CD31) is expressed on cells of the vascular compartment and regulates vascular integrity and immune cell trafficking. Involvement of PECAM-1 in MS pathogenesis has been suggested by the detection of increased levels of soluble PECAM-1 (sPECAM-1) in the serum and CSF of MS patients. Here, we report profound upregulation of cell-bound PECAM-1 in initial (pre-phagocytic) white matter as well as active cortical gray matter MS lesions. Using a human in vitro BBB model we observed that PECAM-1 is not essential for the transmigration of human CD4+ T-cell subsets (Th1, Th1*, Th2, and Th17) across the BBB. Employing an additional in vitro BBB model based on primary mouse brain microvascular endothelial cells (pMBMECs) we show that the lack of endothelial PECAM-1 impairs BBB properties as shown by reduced transendothelial electrical resistance (TEER) and increases permeability for small molecular tracers. Investigating T-cell migration across the BBB under physiological flow by in vitro live cell imaging revealed that absence of PECAM-1 in pMBMECs did not influence arrest, polarization, and crawling of effector/memory CD4+ T cells on the pMBMECs. Absence of endothelial PECAM-1 also did not affect the number of T cells able to cross the pMBMEC monolayer under flow, but surprisingly favored transcellular over paracellular T-cell diapedesis. Taken together, our data demonstrate that PECAM-1 is critically involved in regulating BBB permeability and although not required for T-cell diapedesis itself, its presence or absence influences the cellular route of T-cell diapedesis across the BBB. Upregulated expression of cell-bound PECAM-1 in human MS lesions may thus reflect vascular repair mechanisms aiming to restore BBB integrity and paracellular T-cell migration across the BBB as it occurs during CNS immune surveillance.
Collapse
Affiliation(s)
- Isabella Wimmer
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Silvia Tietz
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zürich,, Zurich, Switzerland
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory, Université d'Artois, Lens, France
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - William A. Muller
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
28
|
Affiliation(s)
- Silvia De Santis
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain
| | - Santiago Canals
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain.
| |
Collapse
|
29
|
Chemical exchange saturation transfer (CEST) as a new method of signal obtainment in magnetic resonance molecular imaging in clinical and research practice. Pol J Radiol 2019; 84:e147-e152. [PMID: 31019609 PMCID: PMC6479148 DOI: 10.5114/pjr.2019.84242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 02/14/2019] [Indexed: 01/04/2023] Open
Abstract
The work describes the physical basis of the chemical exchange saturation transfer (CEST) technique; it presents the beginnings of the implementation of the method and its possible applications. The principles of correct data acquisition and possible solutions used during the design of the CEST sequence are shown. The main problems related to data analysis are indicated, and an example Z-spectrum from in vivo study of the rat brain is introduced. Furthermore, the parameters related to spectrum analyses such as magnetisation transfer asymmetry (MTRasym) and amide proton transfer asymmetry (APTasym) are presented. In the following part, different types of the CEST method often mentioned in the literature are discussed. Subsequently, the possible applications of the CEST method in both clinical and experimental practice are described.
Collapse
|
30
|
The Role of fMRI in the Assessment of Neuroplasticity in MS: A Systematic Review. Neural Plast 2018; 2018:3419871. [PMID: 30693023 PMCID: PMC6332922 DOI: 10.1155/2018/3419871] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/05/2018] [Indexed: 11/17/2022] Open
Abstract
Neuroplasticity, which is the ability of the brain to adapt to internal and external environmental changes, physiologically occurs during growth and in response to damage. The brain's response to damage is of particular interest in multiple sclerosis, a chronic disease characterized by inflammatory and neurodegenerative damage to the central nervous system. Functional MRI (fMRI) is a tool that allows functional changes related to the disease and to its evolution to be studied in vivo. Several studies have shown that abnormal brain recruitment during the execution of a task starts in the early phases of multiple sclerosis. The increased functional activation during a specific task observed has been interpreted mainly as a mechanism of adaptive plasticity designed to contrast the increase in tissue damage. More recent fMRI studies, which have focused on the activity of brain regions at rest, have yielded nonunivocal results, suggesting that changes in functional brain connections represent mechanisms of either adaptive or maladaptive plasticity. The few longitudinal studies available to date on disease evolution have also yielded discrepant results that are likely to depend on the clinical features considered and the length of the follow-up. Lastly, fMRI has been used in interventional studies to investigate plastic changes induced by pharmacological therapy or rehabilitation, though whether such changes represent a surrogate of neuroplasticity remains unclear. The aim of this paper is to systematically review the existing literature in order to provide an overall description of both the neuroplastic process itself and the evolution in the use of fMRI techniques as a means of assessing neuroplasticity. The quantitative and qualitative approach adopted here ensures an objective analysis of published, peer-reviewed research and yields an overview of up-to-date knowledge.
Collapse
|
31
|
Boiziau C, Nikolski M, Mordelet E, Aussudre J, Vargas-Sanchez K, Petry KG. A Peptide Targeting Inflammatory CNS Lesions in the EAE Rat Model of Multiple Sclerosis. Inflammation 2018. [PMID: 29516383 DOI: 10.1007/s10753-018-0748-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis is characterized by inflammatory lesions dispersed throughout the central nervous system (CNS) leading to severe neurological handicap. Demyelination, axonal damage, and blood brain barrier alterations are hallmarks of this pathology, whose precise processes are not fully understood. In the experimental autoimmune encephalomyelitis (EAE) rat model that mimics many features of human multiple sclerosis, the phage display strategy was applied to select peptide ligands targeting inflammatory sites in CNS. Due to the large diversity of sequences after phage display selection, a bioinformatics procedure called "PepTeam" designed to identify peptides mimicking naturally occurring proteins was used, with the goal to predict peptides that were not background noise. We identified a circular peptide CLSTASNSC called "Ph48" as an efficient binder of inflammatory regions of EAE CNS sections including small inflammatory lesions of both white and gray matter. Tested on human brain endothelial cells hCMEC/D3, Ph48 was able to bind efficiently when these cells were activated with IL1β to mimic inflammatory conditions. The peptide is therefore a candidate for further analyses of the molecular alterations in inflammatory lesions.
Collapse
Affiliation(s)
- Claudine Boiziau
- INSERM, UMR 1049, F-33076, Bordeaux, France. .,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France. .,INSERM, UMR 1026, BioTis, F-33 076, Bordeaux, France.
| | - Macha Nikolski
- Univ. Bordeaux, CBiB, F-33076, Bordeaux, France.,CNRS, LaBRI UMR 5800, F-33400, Talence, France
| | - Elodie Mordelet
- INSERM, UMR 1049, F-33076, Bordeaux, France.,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France
| | - Justine Aussudre
- INSERM, UMR 1049, F-33076, Bordeaux, France.,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France
| | - Karina Vargas-Sanchez
- INSERM, UMR 1049, F-33076, Bordeaux, France.,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France.,Biomedical Sciences Research Group, GRINCIBIO, School of Medicine, Universidad Antonio Nariño, Bogotà, Colombia
| | - Klaus G Petry
- INSERM, UMR 1049, F-33076, Bordeaux, France.,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France.,INSERM, UMR1029, F-33076, Bordeaux, France
| |
Collapse
|
32
|
Tietz S, Périnat T, Greene G, Enzmann G, Deutsch U, Adams R, Imhof B, Aurrand-Lions M, Engelhardt B. Lack of junctional adhesion molecule (JAM)-B ameliorates experimental autoimmune encephalomyelitis. Brain Behav Immun 2018; 73:3-20. [PMID: 29920328 DOI: 10.1016/j.bbi.2018.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/11/2018] [Accepted: 06/15/2018] [Indexed: 12/28/2022] Open
Abstract
In multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) autoaggressive CD4+ T cells cross the blood-brain barrier (BBB) and cause neuroinflammation. Therapeutic targeting of CD4+ T-cell trafficking into the CNS by blocking α4-integrins has proven beneficial for the treatment of MS but comes with associated risks, probably due to blocking CD8+ T cell mediated CNS immune surveillance. Our recent observations show that CD8+ T cells also rely on α4β1-integrins to cross the BBB. Besides vascular cell adhesion molecule-1 (VCAM-1), we identified junctional adhesion molecule-B (JAM-B) as a novel vascular α4β1-integrin ligand involved in CD8+ T-cell migration across the BBB. This prompted us to investigate, if JAM-B also mediates CD4+ T-cell migration across the BBB. We first ensured that encephalitogenic T cells can bind to JAM-B in vitro and next compared EAE pathogenesis in JAM-B-/- C57BL/6J mice and their wild-type littermates. Following immunization with MOGaa35-55 peptide, JAM-B-/- mice developed ameliorated EAE compared to their wild-type littermates. At the same time, we isolated higher numbers of CD45+ infiltrating immune cells from the CNS of JAM-B-/- C57BL/6J mice suffering from EAE. Immunofluorescence staining revealed that the majority of CD45+ inflammatory cells accumulated in the leptomeningeal and perivascular spaces of the CNS behind the BBB but do not gain access to the CNS parenchyma. Trapping of CNS inflammatory cells was not due to increased inflammatory cell proliferation. Neither a loss of BBB integrity or BBB polarity potentially affecting local chemokine gradients nor a lack of focal gelatinase activation required for CNS parenchymal immune cell entry across the glia limitans could be detected in JAM-B-/- mice. Lack of a role for JAM-B in the effector phase of EAE was supported by the observation that we did not detect any role for JAM-B in EAE pathogenesis, when EAE was elicited by in vitro activated MOG aa35-55-specific CD4+ effector T cells. On the other hand, we also failed to demonstrate any role of JAM-B in in vivo priming, proliferation or polarization of MOGaa35-55-specific CD4+ T cells in peripheral immune organs. Finally, our study excludes expression of and thus a role for JAM-B on peripheral and CNS infiltrating myeloid cells. Taken together, although endothelial JAM-B is not required for immune cell trafficking across the BBB in EAE, in its absence accumulation of inflammatory cells mainly in CNS leptomeningeal spaces leads to amelioration of EAE.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/metabolism
- CD8-Positive T-Lymphocytes/metabolism
- Cell Movement/physiology
- Central Nervous System/metabolism
- Central Nervous System/physiology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Endothelium, Vascular/metabolism
- Female
- Integrin alpha4beta1/metabolism
- Junctional Adhesion Molecule B/genetics
- Junctional Adhesion Molecule B/metabolism
- Junctional Adhesion Molecule B/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/physiopathology
- Myelin-Oligodendrocyte Glycoprotein/pharmacology
- Myeloid Cells/metabolism
- Myeloid Cells/physiology
- Tight Junctions/metabolism
Collapse
Affiliation(s)
- Silvia Tietz
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Therese Périnat
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gretchen Greene
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Ralf Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Beat Imhof
- Department of Pathology and Immunology, University of Geneva, CMU Geneva, Switzerland
| | - Michel Aurrand-Lions
- Centre de Recherche en Cancerologie de Marseille, INSERM, CNRS, Aix-Marseille University, Marseille, France
| | | |
Collapse
|
33
|
Tommasin S, De Giglio L, Ruggieri S, Petsas N, Giannì C, Pozzilli C, Pantano P. Relation between functional connectivity and disability in multiple sclerosis: a non-linear model. J Neurol 2018; 265:2881-2892. [PMID: 30276520 DOI: 10.1007/s00415-018-9075-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/21/2018] [Accepted: 09/25/2018] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To characterize the relation between brain functional connectivity and disability in patients with multiple sclerosis; to investigate the existence of critical values of both disability and functional connectivity corresponding to exhaustion of functional adaptive mechanisms. METHODS Hundred-and-nineteen patients with no-to-severe disability and 42 healthy subjects were studied via 3T resting state functional MRI. Out of 116 regions extracted from Automated Anatomical Labeling atlas, pairs of regions whose functional connectivity correlated with Expanded Disability Status Score were identified. In patients, mathematical modeling was applied to find the best models describing Expanded-Disability-Status-Score vs structural or functional measures. Functional vs structural models intersecting points were identified. RESULTS Disability had direct linear relation with lesion load (r = 0.40, p < 5E-6), inverse of thalamic volume (r = 0.31 p < 1E-3) and functional connectivity in bi-frontal pairs of regions (r > 0.40, p < 0.04), while being non-linearly associated with functional connectivity in cerebello-temporal and cerebello-frontal pairs of regions (F > 1.73, p < 0.02). Structural vs functional models intersecting points corresponded to Expanded Disability Status Score of 3.0. 85% of patients scoring more than 3.0 showed functional connectivity in cerebello-temporal and cerebello-frontal pairs of regions below confidence intervals (z = [2.28-2.88] 95% CI) measured in healthy subjects. CONCLUSIONS Functional brain connectivity changes may represent mechanisms of adaptation to structural damage and inflammation and may be not always clinically beneficial. Functional connectivity decreases in comparison with structural measure at Expanded Disability Status Score greater than 3.0, which may be critical and indicate exhaustion of compensatory mechanisms.
Collapse
Affiliation(s)
- Silvia Tommasin
- Department of Human Neuroscience, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Laura De Giglio
- Department of Human Neuroscience, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
- Sant'Andrea Hospital, MS Centre, Sapienza University of Rome, Viale di Grottarossa 1035, 00189, Rome, Italy
| | - Serena Ruggieri
- Department of Human Neuroscience, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Nikolaos Petsas
- IRCCS Neuromed, Via Atinense, 18, 86077, Pozzilli, IS, Italy
| | - Costanza Giannì
- Department of Human Neuroscience, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Carlo Pozzilli
- Department of Human Neuroscience, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
- Sant'Andrea Hospital, MS Centre, Sapienza University of Rome, Viale di Grottarossa 1035, 00189, Rome, Italy
| | - Patrizia Pantano
- Department of Human Neuroscience, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy.
- IRCCS Neuromed, Via Atinense, 18, 86077, Pozzilli, IS, Italy.
| |
Collapse
|
34
|
Doty RL, MacGillivray MR, Talab H, Tourbier I, Reish M, Davis S, Cuzzocreo JL, Shepard NT, Pham DL. Balance in multiple sclerosis: relationship to central brain regions. Exp Brain Res 2018; 236:2739-2750. [PMID: 30019234 DOI: 10.1007/s00221-018-5332-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 07/13/2018] [Indexed: 11/30/2022]
Abstract
Dizziness, postural instability, and ataxia are among the most debilitating symptoms of multiple sclerosis (MS), reflecting, in large part, dysfunctional integration of visual, somatosensory, and vestibular sensory cues. However, the role of MS-related supratentorial lesions in producing such symptoms is poorly understood. In this study, motor control test (MCT) and dynamic sensory organization test (SOT) scores of 58 MS patients were compared to those of 72 healthy controls; correlations were determined between the MS scores of 49 patients and lesion volumes within 26 brain regions. Depending upon platform excursion direction and magnitude, MCT latencies, which were longer in MS patients than controls (p < 0.0001), were correlated with lesion volumes in the cortex, medial frontal lobes, temporal lobes, and parietal opercula (r's ranging from 0.20 to 0.39). SOT test scores were also impacted by MS and correlated with lesions in these same brain regions as well as within the superior frontal lobe (r's ranging from - 0.28 to - 0.40). The strongest and most consistent correlations occurred for the most challenging tasks in which incongruent visual and proprioceptive feedback were given. This study demonstrates that supratentorial lesion volumes are associated with quantitative balance measures in MS, in accord with the concept that balance relies upon highly convergent and multimodal neural pathways involving the skin, muscles, joints, eyes, and vestibular system.
Collapse
Affiliation(s)
- Richard L Doty
- Smell and Taste Center, Department of Otorhinolaryngology: Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, 5 Ravdin Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104-4823, USA.
| | - Michael R MacGillivray
- Smell and Taste Center, Department of Otorhinolaryngology: Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, 5 Ravdin Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104-4823, USA
| | - Hussam Talab
- Smell and Taste Center, Department of Otorhinolaryngology: Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, 5 Ravdin Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104-4823, USA
| | - Isabelle Tourbier
- Smell and Taste Center, Department of Otorhinolaryngology: Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, 5 Ravdin Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104-4823, USA
| | - Megan Reish
- Smell and Taste Center, Department of Otorhinolaryngology: Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, 5 Ravdin Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104-4823, USA
| | - Sherrie Davis
- Smell and Taste Center, Department of Otorhinolaryngology: Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, 5 Ravdin Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104-4823, USA
| | | | - Neil T Shepard
- Division of Audiology, Department of Otorhinolaryngology, Mayo Clinic, Rochester, MN, USA
| | - Dzung L Pham
- Center for Neuroscience and Regenerative Medicine, Henry Jackson Foundation, Bethesda, MD, USA
| |
Collapse
|
35
|
Kuo SM. Does Modification of the Large Intestinal Microbiome Contribute to the Anti-Inflammatory Activity of Fermentable Fiber? Curr Dev Nutr 2018; 2:nzx004. [PMID: 30377676 PMCID: PMC6201682 DOI: 10.3945/cdn.117.001180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 11/21/2017] [Indexed: 12/16/2022] Open
Abstract
Fiber is an inadequately understood and insufficiently consumed nutrient. This review examines the possible causal relation between fiber-induced microbiome changes and the anti-inflammatory activity of fiber. To demonstrate the dominant role of fermentable plant fiber in shaping the intestinal microbiome, animal and human fiber-feeding studies are reviewed. Using culture-, PCR-, and sequencing-based microbial analyses, a higher prevalence of Bifidobacterium and Lactobacillus genera was observed from the feeding of different types of fermentable fiber. This finding was reported in studies performed on several host species including human. Health conditions and medications that are linked to intestinal microbial alterations likely also change the nutrient environment of the large intestine. The unique gene clusters of Bifidobacterium and Lactobacillus that enable the catabolism of plant glycans and the ability of Bifidobacterium and Lactobacillus to reduce the colonization of proteobacteria probably contribute to their prevalence in a fiber-rich intestinal environment. The fiber-induced microbiome changes could contribute to the anti-inflammatory activity of fiber. Although most studies did not measure fecal microbial density or total daily fecal microbial output (colon microbial load), limited evidence suggests that the increase in intestinal commensal microbial load plays an important role in the anti-inflammatory activity of fiber. Various probiotic supplements, including Bifidobacterium and Lactobacillus, showed anti-inflammatory activity only in the presence of fiber, which promoted microbial growth as indicated by increasing plasma short-chain fatty acids. Probiotics alone or pure fiber administered under sterile conditions showed no anti-inflammatory activity. The potential mechanisms that could mediate the anti-inflammatory effect of common microbial metabolites are reviewed, but more in vivo trials are needed. Future studies including simultaneous microbial composition and load measurements are also important.
Collapse
Affiliation(s)
- Shiu-Ming Kuo
- Department of Exercise and Nutrition Sciences, University at Buffalo, SUNY, Buffalo, NY
| |
Collapse
|