1
|
Krijnen EA, Lee H, Noteboom S, Chiang FL, Steenwijk MD, Schoonheim MM, Klawiter EC, Huang SY. In vivo evidence for cell body loss in cortical lesions in people with multiple sclerosis. Ann Clin Transl Neurol 2024. [PMID: 39673156 DOI: 10.1002/acn3.52237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 09/28/2024] [Accepted: 10/12/2024] [Indexed: 12/16/2024] Open
Abstract
OBJECTIVE To quantify alterations in soma and neurite density imaging measures within and surrounding cortical lesions in people with multiple sclerosis using in vivo high-gradient diffusion MRI. METHODS In this cross-sectional study, 41 people with multiple sclerosis and 34 age- and sex-matched healthy controls underwent 3 T high-gradient diffusion MRI. Cortical lesions were segmented on artificial intelligence-enabled double inversion recovery images. "Inner" and "outer" perilesional layers were segmented as two expanding shells of 2 mm surrounding a cortical lesion. Intracellular, intra-neurite, and extracellular signal fractions and apparent soma radius were estimated in (peri)lesional and normal-appearing cortex. RESULTS Cortical lesions were present in all people with multiple sclerosis with a median count of 8 [IQR 5-18] and total volume of 0.16 [0.09-0.46 mL]. People with multiple sclerosis (mean 0.27 ± 0.03) showed lower normalized cortical volumes compared to healthy controls (0.30 ± 0.02). Compared to healthy controls (mean 0.58 ± 0.028), normal-appearing cortex in multiple sclerosis (0.57 ± 0.034) showed lower intra-cellular signal fraction. Cortical lesions (0.49 ± 0.089) exhibited lower intra-cellular signal fractions compared to perilesional ("inner": 0.55 ± 0.049, "outer": 0.55 ± 0.039) and normal-appearing cortex, demonstrating a gradation of change. The soma radius varied significantly across cortices, becoming smaller when moving outward from cortical lesions (cortical lesions: 10.38 ± 0.209 μm, "inner" layer: 10.19 ± 0.140 μm, "outer" layer: 10.07 ± 0.149 μm, normal-appearing cortex: 9.99 ± 0.127 μm). INTERPRETATION Cortical cell body loss in multiple sclerosis is most pronounced in cortical lesions and also present in normal-appearing cortex. Gradients of diffusion microstructural alterations moving outward from cortical lesions toward normal-appearing cortex highlight the potential of high-gradient diffusion MRI to identify both focal and diffuse cortical pathology.
Collapse
Affiliation(s)
- Eva A Krijnen
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114, USA
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, 1007 MB, Amsterdam, The Netherlands
| | - Hansol Lee
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 02129, USA
| | - Samantha Noteboom
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, 1007 MB, Amsterdam, The Netherlands
| | - Florence L Chiang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 02129, USA
| | - Martijn D Steenwijk
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, 1007 MB, Amsterdam, The Netherlands
| | - Menno M Schoonheim
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, 1007 MB, Amsterdam, The Netherlands
| | - Eric C Klawiter
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Susie Y Huang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 02129, USA
| |
Collapse
|
2
|
Gadani SP, Singh S, Kim S, Hu J, Smith MD, Calabresi PA, Bhargava P. Spatial transcriptomics of meningeal inflammation reveals inflammatory gene signatures in adjacent brain parenchyma. eLife 2024; 12:RP88414. [PMID: 39475792 PMCID: PMC11524578 DOI: 10.7554/elife.88414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
While modern high efficacy disease modifying therapies have revolutionized the treatment of relapsing-remitting multiple sclerosis, they are less effective at controlling progressive forms of the disease. Meningeal inflammation is a recognized risk factor for cortical gray matter pathology which can result in disabling symptoms such as cognitive impairment and depression, but the mechanisms linking meningeal inflammation and gray matter pathology remain unclear. Here, we performed magnetic resonance imaging (MRI)-guided spatial transcriptomics in a mouse model of autoimmune meningeal inflammation to characterize the transcriptional signature in areas of meningeal inflammation and the underlying brain parenchyma. We found broadly increased activity of inflammatory signaling pathways at sites of meningeal inflammation, but only a subset of these pathways active in the adjacent brain parenchyma. Subclustering of regions adjacent to meningeal inflammation revealed the subset of immune programs induced in brain parenchyma, notably complement signaling and antigen processing/presentation. Trajectory gene and gene set modeling analysis confirmed variable penetration of immune signatures originating from meningeal inflammation into the adjacent brain tissue. This work contributes a valuable data resource to the field, provides the first detailed spatial transcriptomic characterization in a model of meningeal inflammation, and highlights several candidate pathways in the pathogenesis of gray matter pathology.
Collapse
Affiliation(s)
- Sachin P Gadani
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurology, University of PittsburghPittsburghUnited States
| | - Saumitra Singh
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Sophia Kim
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Jingwen Hu
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Matthew D Smith
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Peter A Calabresi
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Solomon Snyder, Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Pavan Bhargava
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
3
|
El Mahdaoui S, Hansen MM, Hansen MB, Hvalkof VH, Søndergaard HB, Mahler MR, Romme Christensen J, Sellebjerg F, von Essen MR. Effects of anti-CD20 therapy on circulating and intrathecal follicular helper T cell subsets in multiple sclerosis. Clin Immunol 2024; 264:110262. [PMID: 38788886 DOI: 10.1016/j.clim.2024.110262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Follicular helper T (Tfh) cells and their interplay with B cells likely contribute to the pathogenesis of relapsing-remitting multiple sclerosis (RRMS). Tfh cells are enriched in cerebrospinal fluid (CSF) in RRMS, but effects of anti-CD20 therapy are unknown. We investigated Tfh cells in controls, untreated and anti-CD20-treated patients with RRMS using flow cytometry. CSF Tfh cells were increased in untreated patients. Compared to paired blood samples, CD25- Tfh cells were enriched in CSF in RRMS, but not in controls. Contrast-enhancing brain MRI lesions and IgG index correlated with CSF CD25- Tfh cell frequency in untreated patients with RRMS. Anti-CD20 therapy reduced the numbers of circulating PD1+ Tfh cells and CD25- Tfh cells, and the frequency of CSF CD25- Tfh cells. The study suggests that CD25- Tfh cells are recruited to the CSF in RRMS, associated with focal inflammation, and are reduced by anti-CD20 therapy.
Collapse
Affiliation(s)
- Sahla El Mahdaoui
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark.
| | - Marie Mathilde Hansen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Malene Bredahl Hansen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Victoria Hyslop Hvalkof
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Helle Bach Søndergaard
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Mie Reith Mahler
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Jeppe Romme Christensen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Marina Rode von Essen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| |
Collapse
|
4
|
Klíčová K, Mareš J, Sobek O, Rous Z, Rous M, Raška M, Hartung HP. Prognostic relevance of the C-X-C motif chemokine ligand 13 and interleukin-8 in predicting the transition from clinically isolated syndrome to multiple sclerosis. Eur J Neurosci 2024; 59:2955-2966. [PMID: 38453679 DOI: 10.1111/ejn.16300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/25/2024] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
The initial phase of multiple sclerosis (MS), often known as clinically isolated syndrome (CIS), is a critical period for identifying individuals at high risk of progressing to full-blown MS and initiating timely treatment. In this study, we aimed to evaluate the prognostic value of C-X-C motif chemokine ligand 13 (CXCL13) and interleukin-8 (IL-8) as potential markers for CIS patients' conversion to MS. Our study encompassed patients with CIS, those with relapsing-remitting MS (RRMS), and control subjects, with sample analysis conducted on both cerebrospinal fluid (CSF) and serum. Patients were categorized into four groups: CIS-CIS (no MS development within 2 years), CIS-RRMS (conversion to RRMS within 2 years), RRMS (already diagnosed), and a control group (CG) with noninflammatory central nervous system disorders. Results showed significantly elevated levels of CXCL13 in CSF across all patient groups compared with the CG (p < 0.0001, Kruskal-Wallis test). Although CXCL13 concentrations were slightly higher in the CIS-RRMS group, statistical significance was not reached. Similarly, significantly higher levels of IL-8 were detected in CSF samples from all patient groups compared with the CG (p < 0.0001, Kruskal-Wallis test). Receiver operating characteristic analysis in the CIS-RRMS group identified both CXCL13 (area under receiver operating characteristic curve = .959) and IL-8 (area under receiver operating characteristic curve = .939) in CSF as significant predictors of CIS to RRMS conversion. In conclusion, our study suggests a trend towards elevated CSF IL-8 and CSF CXCL13 levels in CIS patients progressing to RRMS. These findings emphasize the importance of identifying prognostic markers to guide appropriate treatment strategies for individuals in the early stages of MS.
Collapse
Affiliation(s)
- Kateřina Klíčová
- Department of Neurology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Jan Mareš
- Department of Neurology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Ondřej Sobek
- Laboratory for Cerebrospinal Fluid, Neuroimmunology, Pathology and Special Diagnostics, Topelex, Prague, Czech Republic
| | - Zuzana Rous
- Department of Neurology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Matouš Rous
- Department of Neurology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
| | - Milan Raška
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Hans-Peter Hartung
- Department of Neurology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czech Republic
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
5
|
Jons D, Grut V, Bergström T, Zetterberg H, Biström M, Gunnarsson M, Vrethem M, Brenner N, Butt J, Blennow K, Nilsson S, Kockum I, Olsson T, Waterboer T, Sundström P, Andersen O. Seroreactivity against lytic, latent and possible cross-reactive EBV antigens appears on average 10 years before MS induced preclinical neuroaxonal damage. J Neurol Neurosurg Psychiatry 2024; 95:325-332. [PMID: 37802637 PMCID: PMC10958269 DOI: 10.1136/jnnp-2023-331868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) and presymptomatic axonal injury appear to develop only after an Epstein-Barr virus (EBV) infection. This association remains to be confirmed across a broad preclinical time range, for lytic and latent EBV seroreactivity, and for potential cross-reacting antigens. METHODS We performed a case-control study with 669 individual serum samples obtained before clinical MS onset, identified through cross-linkage with the Swedish MS register. We assayed antibodies against EBV nuclear antigen 1 (EBNA1), viral capsid antigen p18, glycoprotein 350 (gp350), the potential cross-reacting protein anoctamin 2 (ANO2) and the level of sNfL, a marker of axonal injury. RESULTS EBNA1 (latency) seroreactivity increased in the pre-MS group, at 15-20 years before clinical MS onset, followed by gp350 (lytic) seroreactivity (p=0.001-0.009), ANO2 seropositivity appeared shortly after EBNA1-seropositivity in 16.7% of pre-MS cases and 10.0% of controls (p=0.001).With an average lag of almost a decade after EBV, sNfL gradually increased, mainly in the increasing subgroup of seropositive pre-MS cases (p=8.10-5 compared with non-MS controls). Seropositive pre-MS cases reached higher sNfL levels than seronegative pre-MS (p=0.038). In the EBNA1-seropositive pre-MS group, ANO2 seropositive cases had 26% higher sNfL level (p=0.0026). CONCLUSIONS Seroreactivity against latent and lytic EBV antigens, and in a subset ANO2, was detectable on average a decade before the appearance of a gradually increasing axonal injury occurring in the last decade before the onset of clinical MS. These findings strengthen the hypothesis of latent EBV involvement in the pathogenesis of MS.
Collapse
Affiliation(s)
- Daniel Jons
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Viktor Grut
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Tomas Bergström
- Department of Infectious Diseases, Institute of Biomedicine, the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Martin Biström
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Martin Gunnarsson
- Department of Neurology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Magnus Vrethem
- Department of Neurology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Nicole Brenner
- Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Research Program, German Cancer Research Center, Heidelberg, Germany
| | - Julia Butt
- Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Research Program, German Cancer Research Center, Heidelberg, Germany
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Staffan Nilsson
- Mathematical Sciences, Chalmers University of Technology, Göteborg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Goteborg, Sweden
| | - Ingrid Kockum
- Department of Clinical Neuroscience, The Karolinska Neuroimmunology & Multiple Sclerosis Center, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, The Karolinska Neuroimmunology & Multiple Sclerosis Center, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Tim Waterboer
- Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Research Program, German Cancer Research Center, Heidelberg, Germany
| | - Peter Sundström
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Oluf Andersen
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
6
|
Miscioscia A, Treaba CA, Barletta VT, Herranz E, Sloane JA, Barbuti E, Mainero C. White matter paramagnetic rim and non-rim lesions share a periventricular gradient in multiple sclerosis: A 7-T imaging study. Mult Scler 2024; 30:166-176. [PMID: 38279672 PMCID: PMC10922980 DOI: 10.1177/13524585231224681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
BACKGROUND Paramagnetic rim white matter (WM) lesions (PRL) are thought to be a main driver of non-relapsing multiple sclerosis (MS) progression. It is unknown whether cerebrospinal fluid (CSF)-soluble factors diffusing from the ventricles contribute to PRL formation. OBJECTIVE To investigate the distribution of PRL and non-rim brain WM lesions as a function of distance from ventricular CSF, their relationship with cortical lesions, the contribution of lesion phenotype, and localization to neurological disability. METHODS Lesion count and volume of PRL, non-rim WM, leukocortical lesion (LCL), and subpial/intracortical lesions were obtained at 7-T. The brain WM was divided into 1-mm-thick concentric rings radiating from the ventricles to extract PRL and non-rim WM lesion volume from each ring. RESULTS In total, 61 MS patients with ⩾1 PRL were included in the study. Both PRL and non-rim WM lesion volumes were the highest in the periventricular WM and declined with increasing distance from ventricles. A CSF distance-independent association was found between non-rim WM lesions, PRL, and LCL, but not subpial/intracortical lesions. Periventricular non-rim WM lesion volume was the strongest predictor of neurological disability. CONCLUSIONS Non-rim and PRL share a gradient of distribution from the ventricles toward the cortex, suggesting that CSF proximity equally impacts the prevalence of both lesion phenotypes.
Collapse
Affiliation(s)
- Alessandro Miscioscia
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- University of Padova, Padova, Italy
| | - Constantina A. Treaba
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Valeria T. Barletta
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elena Herranz
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jacob A. Sloane
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Elena Barbuti
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Ospedale Sant’Andrea, University La Sapienza, Rome, Italy
| | - Caterina Mainero
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Wen W, Cheng J, Tang Y. Brain perivascular macrophages: current understanding and future prospects. Brain 2024; 147:39-55. [PMID: 37691438 PMCID: PMC10766266 DOI: 10.1093/brain/awad304] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/28/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Brain perivascular macrophages are specialized populations of macrophages that reside in the space around cerebral vessels, such as penetrating arteries and venules. With the help of cutting-edge technologies, such as cell fate mapping and single-cell multi-omics, their multifaceted, pivotal roles in phagocytosis, antigen presentation, vascular integrity maintenance and metabolic regulation have more recently been further revealed under physiological conditions. Accumulating evidence also implies that perivascular macrophages are involved in the pathogenesis of neurodegenerative disease, cerebrovascular dysfunction, autoimmune disease, traumatic brain injury and epilepsy. They can act in either protective or detrimental ways depending on the disease course and stage. However, the underlying mechanisms of perivascular macrophages remain largely unknown. Therefore, we highlight potential future directions in research on perivascular macrophages, including the utilization of genetic mice and novel therapeutic strategies that target these unique immune cells for neuroprotective purposes. In conclusion, this review provides a comprehensive update on the current knowledge of brain perivascular macrophages, shedding light on their pivotal roles in central nervous system health and disease.
Collapse
Affiliation(s)
- Wenjie Wen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Jinping Cheng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| |
Collapse
|
8
|
Hartung HP, Cree BA, Barnett M, Meuth SG, Bar-Or A, Steinman L. Bioavailable central nervous system disease-modifying therapies for multiple sclerosis. Front Immunol 2023; 14:1290666. [PMID: 38162670 PMCID: PMC10755740 DOI: 10.3389/fimmu.2023.1290666] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/09/2023] [Indexed: 01/03/2024] Open
Abstract
Disease-modifying therapies for relapsing multiple sclerosis reduce relapse rates by suppressing peripheral immune cells but have limited efficacy in progressive forms of the disease where cells in the central nervous system play a critical role. To our knowledge, alemtuzumab, fumarates (dimethyl, diroximel, and monomethyl), glatiramer acetates, interferons, mitoxantrone, natalizumab, ocrelizumab, ofatumumab, and teriflunomide are either limited to the periphery or insufficiently studied to confirm direct central nervous system effects in participants with multiple sclerosis. In contrast, cladribine and sphingosine 1-phosphate receptor modulators (fingolimod, ozanimod, ponesimod, and siponimod) are central nervous system-penetrant and could have beneficial direct central nervous system properties.
Collapse
Affiliation(s)
- Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Palacký University Olomouc, Olomouc, Czechia
| | - Bruce A.C. Cree
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, United States
| | - Michael Barnett
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Sven G. Meuth
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Beckman Center for Molecular Medicine, Stanford University Medical Center, Stanford, CA, United States
| |
Collapse
|
9
|
Zong B, Yu F, Zhang X, Zhao W, Li S, Li L. Mechanisms underlying the beneficial effects of physical exercise on multiple sclerosis: focus on immune cells. Front Immunol 2023; 14:1260663. [PMID: 37841264 PMCID: PMC10570846 DOI: 10.3389/fimmu.2023.1260663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Multiple sclerosis (MS) is a prevalent neuroimmunological illness that leads to neurological disability in young adults. Although the etiology of MS is heterogeneous, it is well established that aberrant activity of adaptive and innate immune cells plays a crucial role in its pathogenesis. Several immune cell abnormalities have been described in MS and its animal models, including T lymphocytes, B lymphocytes, dendritic cells, neutrophils, microglia/macrophages, and astrocytes, among others. Physical exercise offers a valuable alternative or adjunctive disease-modifying therapy for MS. A growing body of evidence indicates that exercise may reduce the autoimmune responses triggered by immune cells in MS. This is partially accomplished by restricting the infiltration of peripheral immune cells into the central nervous system (CNS) parenchyma, curbing hyperactivation of immune cells, and facilitating a transition in the balance of immune cells from a pro-inflammatory to an anti-inflammatory state. This review provides a succinct overview of the correlation between physical exercise, immune cells, and MS pathology, and highlights the potential benefits of exercise as a strategy for the prevention and treatment of MS.
Collapse
Affiliation(s)
- Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai, China
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Fengzhi Yu
- College of Physical Education and Health, East China Normal University, Shanghai, China
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- School of Exercise and Health, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
| | - Xiaoyou Zhang
- School of Physical Education, Hubei University, Wuhan, China
| | - Wenrui Zhao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Shichang Li
- College of Physical Education and Health, East China Normal University, Shanghai, China
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Lin Li
- College of Physical Education and Health, East China Normal University, Shanghai, China
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| |
Collapse
|
10
|
Klotz L, Antel J, Kuhlmann T. Inflammation in multiple sclerosis: consequences for remyelination and disease progression. Nat Rev Neurol 2023; 19:305-320. [PMID: 37059811 DOI: 10.1038/s41582-023-00801-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/16/2023]
Abstract
Despite the large number of immunomodulatory or immunosuppressive treatments available to treat relapsing-remitting multiple sclerosis (MS), treatment of the progressive phase of the disease has not yet been achieved. This lack of successful treatment approaches is caused by our poor understanding of the mechanisms driving disease progression. Emerging concepts suggest that a combination of persisting focal and diffuse inflammation within the CNS and a gradual failure of compensatory mechanisms, including remyelination, result in disease progression. Therefore, promotion of remyelination presents a promising intervention approach. However, despite our increasing knowledge regarding the cellular and molecular mechanisms regulating remyelination in animal models, therapeutic increases in remyelination remain an unmet need in MS, which suggests that mechanisms of remyelination and remyelination failure differ fundamentally between humans and demyelinating animal models. New and emerging technologies now allow us to investigate the cellular and molecular mechanisms underlying remyelination failure in human tissue samples in an unprecedented way. The aim of this Review is to summarize our current knowledge regarding mechanisms of remyelination and remyelination failure in MS and in animal models of the disease, identify open questions, challenge existing concepts, and discuss strategies to overcome the translational roadblock in the field of remyelination-promoting therapies.
Collapse
Affiliation(s)
- Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Québec, Canada
| | - Tanja Kuhlmann
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Québec, Canada.
- Institute of Neuropathology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
11
|
Gao D, Zheng CC, Hao JP, Yang CC, Hu CY. Icariin ameliorates behavioral deficits and neuropathology in a mouse model of multiple sclerosis. Brain Res 2023; 1804:148267. [PMID: 36731819 DOI: 10.1016/j.brainres.2023.148267] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023]
Abstract
Multiple sclerosis (MS) is a systemic inflammatory illness of the central nervous system that involves demyelinating lesions in the myelin-rich white matter and pathology in the grey matter. Despite significant advancements in drug research for MS, the disease's complex pathophysiology makes it difficult to treat the progressive forms of the disease. In this study, we identified a natural flavonoid compound icariin (ICA) as a potent effective agent for MS in ameliorating the deterioration of symptoms including the neurological deficit score and the body weight in a murine experimental autoimmune encephalomyelitis (EAE) model. These improvements were associated with decreased demyelination in the corpus callosum and neuron loss in the hippocampus and cortex confirmed by immunohistochemistry analysis. Meanwhile, it was observed that the activation of microglia in cerebral cortex and hippocampus were inhibited followed by the neuroinflammatory cytokines downregulation such as IL-1β, IL-6 and TNF-α after ICA treatment, which was probably attributable to the suppression of microglial NLRP3 inflammasome activation. Additionally, molecular docking also revealed the binding force of ICA to NLRP3 inflammasome protein complexes in vitro. Taken together, our findings have demonstrated that ICA, as pleiotropic agent, prevents EAE-induced MS by improving demyelination and neuron loss, which interferes with the neuroinflammation via microglial NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Dan Gao
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing Engineering Research Center for Nervous System Drugs, Beijing 100053, China
| | - Ceng-Ceng Zheng
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing Engineering Research Center for Nervous System Drugs, Beijing 100053, China
| | - Jin-Ping Hao
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing Engineering Research Center for Nervous System Drugs, Beijing 100053, China
| | - Cui-Cui Yang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing Engineering Research Center for Nervous System Drugs, Beijing 100053, China.
| | - Chao-Ying Hu
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing Engineering Research Center for Nervous System Drugs, Beijing 100053, China; Phase I Clinical Trial Unit, Beijing Ditan Hospital Capital Medical University, Beijing 100015, China.
| |
Collapse
|
12
|
Furman MJ, Meuth SG, Albrecht P, Dietrich M, Blum H, Mares J, Milo R, Hartung HP. B cell targeted therapies in inflammatory autoimmune disease of the central nervous system. Front Immunol 2023; 14:1129906. [PMID: 36969208 PMCID: PMC10034856 DOI: 10.3389/fimmu.2023.1129906] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
Cumulative evidence along several lines indicates that B cells play an important role in the pathological course of multiple sclerosis (MS), neuromyelitisoptica spectrum disorders (NMOSD) and related CNS diseases. This has prompted extensive research in exploring the utility of targeting B cells to contain disease activity in these disorders. In this review, we first recapitulate the development of B cells from their origin in the bone marrow to their migration to the periphery, including the expression of therapy-relevant surface immunoglobulin isotypes. Not only the ability of B cells to produce cytokines and immunoglobulins seems to be essential in driving neuroinflammation, but also their regulatory functions strongly impact pathobiology. We then critically assess studies of B cell depleting therapies, including CD20 and CD19 targeting monoclonal antibodies, as well as the new class of B cell modulating substances, Bruton´s tyrosinekinase (BTK) inhibitors, in MS, NMOSD and MOGAD.
Collapse
Affiliation(s)
- Moritz J. Furman
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Sven G. Meuth
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- Department of Neurology, Maria Hilf Clinic, Moenchengladbach, Germany
| | - Michael Dietrich
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Heike Blum
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Jan Mares
- Department of Neurology, Palacky University in Olomouc, Olomouc, Czechia
| | - Ron Milo
- Department of Neurology, Barzilai Medical Center, Ashkelon, Israel
| | - Hans-Peter Hartung
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- Department of Neurology, Palacky University in Olomouc, Olomouc, Czechia
- Brain and Mind Center, Medical Faculty, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Li G, You Q, Hou X, Zhang S, Du L, Lv Y, Yu L. The effect of exercise on cognitive function in people with multiple sclerosis: a systematic review and meta-analysis of randomized controlled trials. J Neurol 2023; 270:2908-2923. [PMID: 36864256 DOI: 10.1007/s00415-023-11649-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023]
Abstract
OBJECTIVE A growing body of research examining the effect of exercise on cognitive function in people with multiple sclerosis (MS), while findings of available studies were conflicting. We aimed to explore the effect of exercise on cognitive function in MS patients. METHODS For this systematic review and meta-analysis, we searched PubMed, Web of Science, EBSCO, Cochrane, and Scopus electronic databases, through July 18, 2022. Cochrane risk assessment tool was used to evaluate the methodological quality of the included literature. RESULTS Twenty-one studies with a total of 23 experimental groups and 21 control groups met the inclusion criteria. There was a significant effect of exercise on improving cognitive function in MS patients, while the effect size was small (Cohen's d = 0.20, 95% CI 0.06-0.34, p < 0.001, I2 = 39.31%). Subgroup analysis showed that exercise significantly improved memory (Cohen's d = 0.17, 95% CI 0.02-0.33, p = 0.03, I2 = 7.59%). In addition, multicomponent training, exercise conducted 8 weeks and 10 weeks, up to 60 min per session, 3 times or more per week, 180 min or more per week increased cognitive function significantly. Furthermore, a worse basal MS status (defined by the Expanded Disability Status Scale) and an older age were associated with greater improvement in cognitive function. CONCLUSION MS patients are recommended to participate in at least three multicomponent training sessions per week, with each session lasting up to 60 min, and the exercise goal of 180 min per week can be achieved by increasing the frequency of exercise. Exercise lasting 8 or 10 weeks is best for cognitive function improvement. Additionally, a worse basal MS status, or the older the age, the greater effect on cognitive function.
Collapse
Affiliation(s)
- Gen Li
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China.,School of Physical Education & Sports Science, South China Normal University, Guangzhou, China
| | - Qiuping You
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
| | - Xiao Hou
- School of Sport Sciences, Beijing Sport University, Beijing, China
| | - Shiyan Zhang
- Department of Sports Performance, Beijing Sport University, Beijing, China
| | - Liwen Du
- Department of Sports Performance, Beijing Sport University, Beijing, China
| | - Yuanyuan Lv
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China. .,China Institute of Sport and Health Science, Beijing Sport University, Beijing, China.
| | - Laikang Yu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China. .,Department of Sports Performance, Beijing Sport University, Beijing, China.
| |
Collapse
|
14
|
Zamboni S, D'Ambrosio A, Margutti P. Extracellular vesicles as contributors in the pathogenesis of multiple sclerosis. Mult Scler Relat Disord 2023; 71:104554. [PMID: 36842311 DOI: 10.1016/j.msard.2023.104554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/22/2023]
Abstract
Extracellular vesicles (EVs) are a heterogeneous family of extracellular structures bounded by a phospholipid bilayer, released by all cell types in various biological fluids, such as blood and cerebrospinal fluid (CSF), playing important roles in intercellular communication, both locally and systemically. EVs carry and deliver a variety of bioactive molecules (proteins, nucleic acids, lipids and metabolites), conferring epigenetic and phenotypic changes to the recipient cells and thus resulting as important mediators of both homeostasis and pathogenesis. In neurological diseases, such as multiple sclerosis (MS), the EV ability to cross Blood-Brain Barrier (BBB), moving from central nervous system (CNS) to the peripheral circulation and vice versa, has increased the interest in EV study in the neurological field. In the present review, we will provide an overview of the recent advances made in understanding the pathogenic role of EVs regarding the immune response, the BBB dysfunction and the CNS inflammatory processes.
Collapse
Affiliation(s)
- Silvia Zamboni
- Department of Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | | | - Paola Margutti
- Department of Neurosciences, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
15
|
Rosenstein I, Axelsson M, Novakova L, Rasch S, Blennow K, Zetterberg H, Lycke J. High levels of kappa free light chain synthesis predict cognitive decline in relapsing-remitting multiple sclerosis. Front Immunol 2023; 14:1106028. [PMID: 36742305 PMCID: PMC9896185 DOI: 10.3389/fimmu.2023.1106028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
Background Evolving evidence suggests that measurement of cerebrospinal fluid (CSF) kappa free light chain (KFLC) synthesis has high diagnostic sensitivity and specificity for multiple sclerosis (MS), but its prognostic ability is less investigated. The usefulness of KFLC in predicting cognitive impairment (CI) is still unknown. Methods In a monocentric longitudinal retrospecitve cohort study, KFLC-index ([CSF KFLC/serum KFLC]/[CSF albumin/serum albumin]) measured by latex-enhanced immunonephelometry was prospectively determined as part of the diagnostic workup in patients with early relapsing-remitting MS (RRMS, n=77). The ability of KFLC-index to predict information processing speed (IPS) worsening as assessed with the symbol digit modalities test (SDMT) was investigated in univariable and multivariable models. Results In patients with KFLC-index>100 (n=31), 11 subjects (35.5%) showed reduced SDMT scores by ≥8 points at follow-up (mean follow-up time 7.3 ± 2.6 years), compared with their baseline scores (p=0.01). Baseline KFLC-index>100 was strongly associated with a higher hazard of SDMT score reduction at follow-up (adjusted hazard ratio 10.5, 95% confidence interval 2.2-50.8, p=0.003; median time to SDMT reduction 7 years). Conclusion Intrathecal KFLC synthesis has become an attractive diagnostic tool for MS. We show for the first time that in a real-world setting of early RRMS, KFLC-index predicted cognitive decline. Whether this predictive ability of KFLC-index also concerns other cognitive domains than IPS, warrants further investigations.
Collapse
Affiliation(s)
- Igal Rosenstein
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,*Correspondence: Igal Rosenstein,
| | - Markus Axelsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lenka Novakova
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sofia Rasch
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden,United Kingdom (UK) Dementia Research Institute at University College London (UCL), London, United Kingdom,Department of Neurodegenerative Disease, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom,Hong Kong Centre for Neurodegenerative Diseases, Hong Kong SAR, China,Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
16
|
Chemokines induced by PEDV infection and chemotactic effects on monocyte, T and B cells. Vet Microbiol 2022; 275:109599. [DOI: 10.1016/j.vetmic.2022.109599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
|
17
|
Cao T, Wang Z, Zhu X. The Immunomodulatory Functions of BTK Inhibition in the Central Nervous System. J Inflamm Res 2022; 15:6427-6438. [DOI: 10.2147/jir.s389958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
|