Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Publication year range
1.
Nat Methods ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890427

ABSTRACT

The dry mass and the orientation of biomolecules can be imaged without a label by measuring their permittivity tensor (PT), which describes how biomolecules affect the phase and polarization of light. Three-dimensional (3D) imaging of PT has been challenging. We present a label-free computational microscopy technique, PT imaging (PTI), for the 3D measurement of PT. PTI encodes the invisible PT into images using oblique illumination, polarization-sensitive detection and volumetric sampling. PT is decoded from the data with a vectorial imaging model and a multi-channel inverse algorithm, assuming uniaxial symmetry in each voxel. We demonstrate high-resolution imaging of PT of isotropic beads, anisotropic glass targets, mouse brain tissue, infected cells and histology slides. PTI outperforms previous label-free imaging techniques such as vector tomography, ptychography and light-field imaging in resolving the 3D orientation and symmetry of organelles, cells and tissue. We provide open-source software and modular hardware to enable the adoption of the method.

2.
Nat Immunol ; 14(11): 1166-72, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24076635

ABSTRACT

Sphingosine 1-phosphate (S1P) signaling regulates lymphocyte egress from lymphoid organs into systemic circulation. The sphingosine phosphate receptor 1 (S1P1) agonist FTY-720 (Gilenya) arrests immune trafficking and prevents multiple sclerosis (MS) relapses. However, alternative mechanisms of S1P-S1P1 signaling have been reported. Phosphoproteomic analysis of MS brain lesions revealed S1P1 phosphorylation on S351, a residue crucial for receptor internalization. Mutant mice harboring an S1pr1 gene encoding phosphorylation-deficient receptors (S1P1(S5A)) developed severe experimental autoimmune encephalomyelitis (EAE) due to autoimmunity mediated by interleukin 17 (IL-17)-producing helper T cells (TH17 cells) in the peripheral immune and nervous system. S1P1 directly activated the Jak-STAT3 signal-transduction pathway via IL-6. Impaired S1P1 phosphorylation enhances TH17 polarization and exacerbates autoimmune neuroinflammation. These mechanisms may be pathogenic in MS.


Subject(s)
Brain/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interleukin-17/metabolism , Lysophospholipids/metabolism , Multiple Sclerosis/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction/immunology , Sphingosine/analogs & derivatives , Animals , Autopsy , Brain/immunology , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Humans , Inflammation , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Janus Kinases/genetics , Janus Kinases/immunology , Janus Kinases/metabolism , Lysophospholipids/immunology , Mice , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Phosphorylation , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Sphingosine/immunology , Sphingosine/metabolism , Th17 Cells
3.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33653955

ABSTRACT

Pericytes regulate the development of organ-specific characteristics of the brain vasculature such as the blood-brain barrier (BBB) and astrocytic end-feet. Whether pericytes are involved in the control of leukocyte trafficking in the adult central nervous system (CNS), a process tightly regulated by CNS vasculature, remains elusive. Using adult pericyte-deficient mice (Pdgfbret/ret ), we show that pericytes limit leukocyte infiltration into the CNS during homeostasis and autoimmune neuroinflammation. The permissiveness of the vasculature toward leukocyte trafficking in Pdgfbret/ret mice inversely correlates with vessel pericyte coverage. Upon induction of experimental autoimmune encephalomyelitis (EAE), pericyte-deficient mice die of severe atypical EAE, which can be reversed with fingolimod, indicating that the mortality is due to the massive influx of immune cells into the brain. Additionally, administration of anti-VCAM-1 and anti-ICAM-1 antibodies reduces leukocyte infiltration and diminishes the severity of atypical EAE symptoms of Pdgfbret/ret mice, indicating that the proinflammatory endothelium due to absence of pericytes facilitates exaggerated neuroinflammation. Furthermore, we show that the presence of myelin peptide-specific peripheral T cells in Pdgfbret/ret ;2D2tg mice leads to the development of spontaneous neurological symptoms paralleled by the massive influx of leukocytes into the brain. These findings indicate that intrinsic changes within brain vasculature can promote the development of a neuroinflammatory disorder.


Subject(s)
Blood-Brain Barrier/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Homeostasis/immunology , Leukocytes/immunology , Pericytes/immunology , Animals , Blood-Brain Barrier/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Homeostasis/genetics , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Leukocytes/pathology , Mice , Mice, Transgenic , Pericytes/pathology , Proto-Oncogene Proteins c-sis/deficiency , Proto-Oncogene Proteins c-sis/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
4.
Cell Mol Neurobiol ; 43(3): 1219-1236, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35917044

ABSTRACT

Multiple sclerosis (MS) is an inflammatory-demyelinating disease of the central nervous system (CNS) mediated by aberrant auto-reactive immune responses. The current immune-modulatory therapies are unable to protect and repair immune-mediated neural tissue damage. One of the therapeutic targets in MS is the sphingosine-1-phosphate (S1P) pathway which signals via sphingosine-1-phosphate receptors 1-5 (S1P1-5). S1P receptors are expressed predominantly on immune and CNS cells. Considering the potential neuroprotective properties of S1P signaling, we utilized S1P1-GFP (Green fluorescent protein) reporter mice in the cuprizone-induced demyelination model to investigate in vivo S1P - S1P1 signaling in the CNS. We observed S1P1 signaling in a subset of neural stem cells in the subventricular zone (SVZ) during demyelination. During remyelination, S1P1 signaling is expressed in oligodendrocyte progenitor cells in the SVZ and mature oligodendrocytes in the medial corpus callosum (MCC). In the cuprizone model, we did not observe S1P1 signaling in neurons and astrocytes. We also observed ß-arrestin-dependent S1P1 signaling in lymphocytes during demyelination and CNS inflammation. Our findings reveal ß-arrestin-dependent S1P1 signaling in oligodendrocyte lineage cells implying a role of S1P1 signaling in remyelination.


Subject(s)
Multiple Sclerosis , Remyelination , Mice , Animals , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/therapeutic use , Cuprizone , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/therapeutic use , Central Nervous System/metabolism , Multiple Sclerosis/metabolism , Oligodendroglia/metabolism , beta-Arrestins/metabolism , beta-Arrestins/therapeutic use , Mice, Inbred C57BL
5.
Nature ; 523(7560): 342-6, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26053123

ABSTRACT

Lipid mediators influence immunity in myriad ways. For example, circulating sphingosine-1-phosphate (S1P) is a key regulator of lymphocyte egress. Although the majority of plasma S1P is bound to apolipoprotein M (ApoM) in the high-density lipoprotein (HDL) particle, the immunological functions of the ApoM-S1P complex are unknown. Here we show that ApoM-S1P is dispensable for lymphocyte trafficking yet restrains lymphopoiesis by activating the S1P1 receptor on bone marrow lymphocyte progenitors. Mice that lacked ApoM (Apom(-/-)) had increased proliferation of Lin(-) Sca-1(+) cKit(+) haematopoietic progenitor cells (LSKs) and common lymphoid progenitors (CLPs) in bone marrow. Pharmacological activation or genetic overexpression of S1P1 suppressed LSK and CLP cell proliferation in vivo. ApoM was stably associated with bone marrow CLPs, which showed active S1P1 signalling in vivo. Moreover, ApoM-bound S1P, but not albumin-bound S1P, inhibited lymphopoiesis in vitro. Upon immune stimulation, Apom(-/-) mice developed more severe experimental autoimmune encephalomyelitis, characterized by increased lymphocytes in the central nervous system and breakdown of the blood-brain barrier. Thus, the ApoM-S1P-S1P1 signalling axis restrains the lymphocyte compartment and, subsequently, adaptive immune responses. Unique biological functions imparted by specific S1P chaperones could be exploited for novel therapeutic opportunities.


Subject(s)
Apolipoproteins/metabolism , Central Nervous System/pathology , Lipoproteins, HDL/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Lymphopoiesis , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Apolipoproteins/deficiency , Apolipoproteins/genetics , Apolipoproteins M , Blood-Brain Barrier/pathology , Cell Movement , Cell Proliferation/genetics , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Fingolimod Hydrochloride/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lymphocytes/immunology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lysophospholipids/agonists , Lysophospholipids/blood , Lysophospholipids/genetics , Male , Mice , Mice, Inbred C57BL , Protein Binding , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/agonists , Sphingosine/blood , Sphingosine/genetics , Sphingosine/metabolism
6.
Proc Natl Acad Sci U S A ; 114(9): E1745-E1754, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28196893

ABSTRACT

The small heat shock protein αB-crystallin (CRYAB) has been implicated in multiple sclerosis (MS) pathogenesis. Earlier studies have indicated that CRYAB inhibits inflammation and attenuates clinical disease when administered in the experimental autoimmune encephalomyelitis model of MS. In this study, we evaluated the role of CRYAB in primary demyelinating events. Using the cuprizone model of demyelination, a noninflammatory model that allows the analysis of glial responses in MS, we show that endogenous CRYAB expression is associated with increased severity of demyelination. Moreover, we demonstrate a strong correlation between the expression of CRYAB and the extent of reactive astrogliosis in demyelinating areas and in in vitro assays. In addition, we reveal that CRYAB is differentially phosphorylated in astrocytes in active demyelinating MS lesions, as well as in cuprizone-induced lesions, and that this phosphorylation is required for the reactive astrocyte response associated with demyelination. Furthermore, taking a proteomics approach to identify proteins that are bound by the phosphorylated forms of CRYAB in primary cultured astrocytes, we show that there is clear differential binding of protein targets due to the specific phosphorylation of CRYAB. Subsequent Ingenuity Pathway Analysis of these targets reveals implications for intracellular pathways and biological processes that could be affected by these modifications. Together, these findings demonstrate that astrocytes play a pivotal role in demyelination, making them a potential target for therapeutic intervention, and that phosphorylation of CRYAB is a key factor supporting the pathogenic response of astrocytes to oligodendrocyte injury.


Subject(s)
Astrocytes/metabolism , Demyelinating Diseases/metabolism , Phosphorylation/physiology , alpha-Crystallin B Chain/metabolism , Animals , Astrocytes/drug effects , Cuprizone/pharmacology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/chemically induced , Multiple Sclerosis/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Phosphorylation/drug effects
7.
J Autoimmun ; 105: 102290, 2019 12.
Article in English | MEDLINE | ID: mdl-31202617

ABSTRACT

The critical role of sphingosine-1-phosphate (S1P) signaling in lymphocyte trafficking is well recognized, however, the contribution of myeloid cell-S1P signaling in neuroimmunity is less well understood. We previously reported that C57BL/6J mice harboring phosphorylation defective S1P receptor 1 (S1P1) (with mutated serines in the carboxyl terminus, leading to impaired receptor internalization) [S1P1(S5A)] developed severe, TH17-dominant experimental autoimmune encephalomyelitis. In this study, we demonstrate that S1P1-mediated TH17 polarization is not an intrinsic T cell effect, but dependent on sustained S1P1 signaling in myeloid cells. First, utilizing the S1P1(S5A) mice in the EAE model, we observed that S1P1 activated and enhanced antigen presentation function in myeloid cells. Second, sequential phosphorylation of STAT3 occurred in dendritic cells, monocytes, and macrophages/microglia during neuroinflammation. Third, we show that pro-inflammatory (CD45hiCD11b+Ly6Chi) monocytes contribute to TH17 differentiation and neuroinflammation by regulating IL-6 expression. Finally, results from experiments utilizing myeloid cell-specific S1P1 overexpression (S1pr1f/stop/f:LysMCre) mice demonstrate that myeloid cell S1P1 directly contributes to severity of neuroinflammation. These findings reveal the critical contribution of myeloid-S1P1 signaling in CNS autoimmunity.


Subject(s)
Autoimmunity/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Myeloid Cells/immunology , Sphingosine-1-Phosphate Receptors/immunology , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/metabolism , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
9.
Proteomics ; 17(6)2017 03.
Article in English | MEDLINE | ID: mdl-28191734

ABSTRACT

In order to gain mechanistic insights into multiple sclerosis (MS) pathogenesis, we utilized a multi-dimensional approach to test the hypothesis that mutations in myelin proteins lead to immune activation and central nervous system autoimmunity in MS. Mass spectrometry-based proteomic analysis of human MS brain lesions revealed seven unique mutations of PLP1; a key myelin protein that is known to be destroyed in MS. Surprisingly, in-depth genomic analysis of two MS patients at the genomic DNA and mRNA confirmed mutated PLP1 in RNA, but not in the genomic DNA. Quantification of wild type and mutant PLP RNA levels by qPCR further validated the presence of mutant PLP RNA in the MS patients. To seek evidence linking mutations in abundant myelin proteins and immune-mediated destruction of myelin, specific immune response against mutant PLP1 in MS patients was examined. Thus, we have designed paired, wild type and mutant peptide microarrays, and examined antibody response to multiple mutated PLP1 in sera from MS patients. Consistent with the idea of different patients exhibiting unique mutation profiles, we found that 13 out of 20 MS patients showed antibody responses against specific but not against all the mutant-PLP1 peptides. Interestingly, we found mutant PLP-directed antibody response against specific mutant peptides in the sera of pre-MS controls. The results from integrative proteomic, genomic, and immune analyses reveal a possible mechanism of mutation-driven pathogenesis in human MS. The study also highlights the need for integrative genomic and proteomic analyses for uncovering pathogenic mechanisms of human diseases.


Subject(s)
Allergy and Immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Mutation/genetics , Myelin Proteolipid Protein/genetics , Proteomics/methods , Translational Research, Biomedical/methods , Amino Acid Sequence , Antibodies/immunology , Female , Humans , Models, Biological , Myelin Proteolipid Protein/chemistry
10.
Stem Cells ; 34(7): 1812-25, 2016 07.
Article in English | MEDLINE | ID: mdl-26989838

ABSTRACT

Cellular transplantation strategies utilizing intraspinal injection of mesenchymal progenitor cells (MPCs) have been reported as beneficial for spinal cord injuries. However, intraspinal injection is not only technically challenging, but requires invasive surgical procedures for patients. Therefore, we investigated the feasibility and potential benefits of noninvasive intravenous injection of MPCs in two models of cervical spinal cord injury, unilateral C5 contusion and complete unilateral C5 hemisection. MPCs isolated from green fluorescence protein (GFP)-luciferase transgenic mice compact bone (1 × 10(6) cells), or vehicle Hank's Buffered Saline Solution (HBSS), were intravenously injected via the tail vein at D1, D3, D7, D10, or D14. Transplanted MPCs were tracked via bioluminescence imaging. Live in vivo imaging data showed that intravenously injected MPCs accumulate in the lungs, confirmed by postmortem bioluminescence signal-irrespective of the time of injection or injury model. The results showed a rapid, positive modulation of the inflammatory response providing protection to the injured spinal cord tissue. Histological processing of the lungs showed GFP(+) cells evenly distributed around the alveoli. We propose that injected cells can act as cellular target decoys to an immune system primed by injury, thereby lessening the inflammatory response at the injury site. We also propose that intravenous injected MPCs modulate the immune system via the lungs through secreted immune mediators or contact interaction with peripheral organs. In conclusion, the timing of intravenous injection of MPCs is key to the success for improving function and tissue preservation following cervical spinal cord injury. Stem Cells 2016;34:1812-1825.


Subject(s)
Cervical Vertebrae/pathology , Lung/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Spinal Cord Injuries/therapy , Administration, Intravenous , Animals , Axons/pathology , Behavior, Animal , CD11b Antigen/metabolism , Chondroitin Sulfates/metabolism , Contusions , Female , Immunohistochemistry , Luminescent Measurements , Mice , Neovascularization, Physiologic , Neuroglia/pathology , Spinal Cord Injuries/pathology , Tissue Distribution , Treatment Outcome
11.
Immunology ; 142(3): 347-53, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24597601

ABSTRACT

Sphingosine-1-phosphate (S1P) is a lipid second messenger that signals via five G protein-coupled receptors (S1P1-5 ). S1P receptor (S1PR) signalling is associated with a wide variety of physiological processes including lymphocyte biology, their recirculation and determination of T-cell phenotypes. The effect of FTY720 (Fingolimod, Gilenya™) to regulate lymphocyte egress and to ameliorate paralysis in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis led to the use of FTY720 as a first-line oral agent for treatment of relapsing-remitting multiple sclerosis. However, a significant body of research suggests that S1P signalling may participate in diverse immune regulatory functions other than lymphocyte trafficking. This review article discusses the current knowledge of S1P signalling in the fate and function of T regulatory, T helper type 17 and memory T cells in health and disease.


Subject(s)
Receptors, Lysosphingolipid/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Humans , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/cytology
12.
Nature ; 451(7182): 1076-81, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18278032

ABSTRACT

Understanding the neuropathology of multiple sclerosis (MS) is essential for improved therapies. Therefore, identification of targets specific to pathological types of MS may have therapeutic benefits. Here we identify, by laser-capture microdissection and proteomics, proteins unique to three major types of MS lesions: acute plaque, chronic active plaque and chronic plaque. Comparative proteomic profiles identified tissue factor and protein C inhibitor within chronic active plaque samples, suggesting dysregulation of molecules associated with coagulation. In vivo administration of hirudin or recombinant activated protein C reduced disease severity in experimental autoimmune encephalomyelitis and suppressed Th1 and Th17 cytokines in astrocytes and immune cells. Administration of mutant forms of recombinant activated protein C showed that both its anticoagulant and its signalling functions were essential for optimal amelioration of experimental autoimmune encephalomyelitis. A proteomic approach illuminated potential therapeutic targets selective for specific pathological stages of MS and implicated participation of the coagulation cascade.


Subject(s)
Gene Expression Profiling , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Proteomics , Adult , Animals , Blood Coagulation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Middle Aged , Multiple Sclerosis/classification , Multiple Sclerosis/drug therapy , Protein C/genetics , Protein C/metabolism , Protein C/pharmacology , Th1 Cells/immunology , Th2 Cells/immunology , Thrombin/antagonists & inhibitors , Thrombin/metabolism
13.
Neurology ; 102(1): e207965, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38165361

ABSTRACT

BACKGROUND AND OBJECTIVES: Neuromyelitis optica spectrum disorder (NMOSD) is a chronic CNS demyelinating autoimmune disorder targeting the astrocyte antigen aquaporin-4 (AQP4), typically presenting with optic neuritis, transverse myelitis, and brain syndromes. Cognitive dysfunction (CD) in NMOSD is under-recognized and poorly understood. The purpose of this study was to evaluate the prevalence and clinical variables associated with CD in NMOSD. METHODS: This observational retrospective study with longitudinal follow-up describes a clinical cohort seen in the Collaborative International Research in Clinical and Longitudinal Experience Study in NMOSD. Serial Montreal Cognitive Assessments (MoCAs) were performed upon enrollment and at 6-month intervals to evaluate longitudinal cognitive function relative to demographic and disease-related factors. We used 2-tailed t test, analysis of variance, the χ2 test, linear regression for univariable and adjusted analyses and simultaneous linear regression and mixed-effects model for multivariable analyses. RESULTS: Thirty-four percent (75/219) of patients met criteria for CD (MoCA <26); 29% (64/219) showed mild dysfunction (MoCA 20-26/30), and 5% (11/219) showed moderate (MoCA <20/30) dysfunction. Patients with less neurologic disability and lower pain scores had higher MoCA scores (95% CI 0.24-0.65 and 95% CI 0.09-0.42, respectively). Patients with at least high school education scored higher on the MoCA (95% CI 2.2-5). When comparing patients dichotomized for CD, patients never on rituximab scored higher than patients only treated with rituximab (p < 0.029). There was no significant association between annualized relapse rate, age, sex, disease duration, AQP4 serostatus or brain lesions, and CD. CD was more pronounced among Black than White patients (95% CI -2.7 to -0.7). Multivariable analysis of serial MoCA did not indicate change (p = 0.715). Descriptive analysis of serial MoCA showed 30% (45/150) of patients with worsening MoCA performance had impaired language and verbal recall. DISCUSSION: To our knowledge, this is the largest study of diverse cohort to investigate CD in patients with NMOSD. Our findings demonstrate 34% of patients with NMOSD experience mild-to-moderate CD, while 30% of patients demonstrated decline on serial testing. The substantial prevalence of CD in this pilot report highlights the need for improved and validated screening tools and comprehensive measures to investigate CD in NMOSD.


Subject(s)
Cognitive Dysfunction , Neuromyelitis Optica , Humans , Neuromyelitis Optica/complications , Neuromyelitis Optica/epidemiology , Prevalence , Retrospective Studies , Rituximab , Neoplasm Recurrence, Local , Cognitive Dysfunction/epidemiology , Aquaporin 4
14.
Blood Adv ; 8(6): 1474-1486, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38295285

ABSTRACT

ABSTRACT: CD19 chimeric antigen receptor (CAR) T-cell therapy has proven highly effective for treating relapsed/refractory mantle cell lymphoma (MCL). However, immune effector cell-associated neurotoxicity syndrome (ICANS) remains a significant concern. This study aimed to evaluate the clinical, radiological, and laboratory correlatives associated with ICANS development after CD19 CAR T-cell therapy in patients with MCL. All patients (N = 26) who received standard-of-care brexucabtagene autoleucel until July 2022 at our institution were evaluated. Laboratory and radiographic correlatives including brain magnetic resonance imaging (MRI) and electroencephalogram (EEG) were evaluated to determine the clinical impact of ICANS. Seventeen (65%) patients experienced ICANS after treatment, with a median onset on day 6. Ten (38%) patients experienced severe (grade ≥3) ICANS. All patients with ICANS had antecedent cytokine release syndrome (CRS), but no correlation was observed between ICANS severity and CRS grade. Overall, 92% of EEGs revealed interictal changes; no patients experienced frank seizures because of ICANS. In total, 86% of patients with severe ICANS with postinfusion brain MRIs demonstrated acute neuroimaging findings not seen on pretreatment MRI. Severe ICANS was also associated with higher rates of cytopenia, coagulopathy, increased cumulative steroid exposure, and prolonged hospitalization. However, severe ICANS did not affect treatment outcomes of patients with MCL. Severe ICANS is frequently associated with a range of postinfusion brain MRI changes and abnormal EEG findings. Longer hospitalization was observed in patients with severe ICANS, especially those with abnormal acute MRI or EEG findings, but there was no discernible impact on overall treatment response and survival.


Subject(s)
Lymphoma, Mantle-Cell , Neurotoxicity Syndromes , Humans , Adult , Lymphoma, Mantle-Cell/therapy , Immunotherapy, Adoptive/adverse effects , Adaptor Proteins, Signal Transducing , Antigens, CD19 , Brain , Cytokine Release Syndrome
15.
Mult Scler ; 19(1): 5-14, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23303879

ABSTRACT

Four questions were posed about multiple sclerosis (MS) at the 2011 Charcot Lecture, Oct. 22, 2011. 1. The Male/Female Disparity: Why are women developing MS so much more frequently than men? 2. Neuronal and Glial Protection: Are there guardian molecules that protect the nervous system in MS? 3. Predictive Medicine: With all the approved drugs, how can we rationally decide which one to use? 4. The Precise Scalpel vs. the Big Hammer for Therapy: Is antigen-specific therapy for demyelinating disease possible? To emphasize how our views on the pathogenesis and treatment of MS are evolving, and given the location of the talk in Amsterdam, Piet Mondrian's progressive interpretations of trees serve as a heuristic.


Subject(s)
Multiple Sclerosis , Humans
16.
Mult Scler ; 18(4): 398-408, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22343184

ABSTRACT

BACKGROUND: The pathology of neuromyelitis optica (NMO), in contrast to multiple sclerosis, comprises granulocyte infiltrates along extensive lengths of spinal cord, as well as optic nerve. Furthermore, IFN-ß treatment worsens NMO. We recently found that experimental autoimmune encephalomyelitis (EAE) induced with Th17 cells is exacerbated by IFN-ß, in contrast to disease induced with Th1 where treatment attenuated symptoms. OBJECTIVE: This study demonstrates the similarities between NMO and Th17 EAE and how neutrophils mediate pathology in Th17 disease. METHODS: Levels of blood biomarkers in NMO were assessed by Luminex and ELISA. Effects of IFN-ß on neutrophils were assessed by culture assays and immunofluorescence. EAE was induced by transfer of myelin-specific Th1 or Th17 cells and treated with Sivelestat sodium hydrate, a neutrophil elastase inhibitor. RESULTS: We show Th17 cytokines, granulocyte chemokines, type 1 interferon and neutrophil elastase are elevated in patients with definitive NMO. In culture, we find that IFN-ß stimulates neutrophils to release neutrophil elastase. In Th17 EAE, we demonstrate neutrophilic infiltration in the optic nerve and spinal cord which was not present in Th1 EAE. Blockade of neutrophil elastase with Sivelestat had efficacy in Th17 EAE but not Th1 EAE. CONCLUSIONS: The similarities between Th17 EAE and NMO indicate that this model represents several aspects of NMO. Neutrophils are critical in the pathologies of both Th17-EAE and NMO, and therefore blockade of neutrophil elastase is a promising target in treating NMO.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Glycine/analogs & derivatives , Neuromyelitis Optica/drug therapy , Pancreatic Elastase/antagonists & inhibitors , Serine Proteinase Inhibitors/therapeutic use , Sulfonamides/therapeutic use , Adolescent , Adult , Animals , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Glycine/therapeutic use , Glycoproteins/immunology , Humans , Interferon-beta/pharmacology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myelin-Oligodendrocyte Glycoprotein , Neuromyelitis Optica/immunology , Neuromyelitis Optica/pathology , Peptide Fragments/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Young Adult
17.
Brain ; 134(Pt 9): 2703-21, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21908875

ABSTRACT

High-throughput technologies have led to advances in the recognition of disease pathways and their underlying mechanisms. To investigate the impact of micro-RNAs on the disease process in multiple sclerosis, a prototypic inflammatory neurological disorder, we examined cerebral white matter from patients with or without the disease by micro-RNA profiling, together with confirmatory reverse transcription-polymerase chain reaction analysis, immunoblotting and gas chromatography-mass spectrometry. These observations were verified using the in vivo multiple sclerosis model, experimental autoimmune encephalomyelitis. Brains of patients with or without multiple sclerosis demonstrated differential expression of multiple micro-RNAs, but expression of three neurosteroid synthesis enzyme-specific micro-RNAs (miR-338, miR-155 and miR-491) showed a bias towards induction in patients with multiple sclerosis (P < 0.05). Analysis of the neurosteroidogenic pathways targeted by micro-RNAs revealed suppression of enzyme transcript and protein levels in the white matter of patients with multiple sclerosis (P < 0.05). This was confirmed by firefly/Renilla luciferase micro-RNA target knockdown experiments (P < 0.05) and detection of specific micro-RNAs by in situ hybridization in the brains of patients with or without multiple sclerosis. Levels of important neurosteroids, including allopregnanolone, were suppressed in the white matter of patients with multiple sclerosis (P < 0.05). Induction of the murine micro-RNAs, miR-338 and miR-155, accompanied by diminished expression of neurosteroidogenic enzymes and allopregnanolone, was also observed in the brains of mice with experimental autoimmune encephalomyelitis (P < 0.05). Allopregnanolone treatment of the experimental autoimmune encephalomyelitis mouse model limited the associated neuropathology, including neuroinflammation, myelin and axonal injury and reduced neurobehavioral deficits (P < 0.05). These multi-platform studies point to impaired neurosteroidogenesis in both multiple sclerosis and experimental autoimmune encephalomyelitis. The findings also indicate that allopregnanolone and perhaps other neurosteroid-like compounds might represent potential biomarkers or therapies for multiple sclerosis.


Subject(s)
MicroRNAs/metabolism , Multiple Sclerosis/metabolism , Neurotransmitter Agents/biosynthesis , 20-Hydroxysteroid Dehydrogenases/genetics , 20-Hydroxysteroid Dehydrogenases/metabolism , Anesthetics/pharmacology , Anesthetics/therapeutic use , Animals , Cells, Cultured , Computational Biology , Encephalitis/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Neurotransmitter Agents/genetics , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/physiology , Pregnanolone/pharmacology , Pregnanolone/therapeutic use , Rats , Rats, Sprague-Dawley
18.
Proc Natl Acad Sci U S A ; 106(35): 14948-53, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19706421

ABSTRACT

The renin-angiotensin-aldosterone system (RAAS) is a major regulator of blood pressure. The octapeptide angiotensin II (AII) is proteolytically processed from the decapeptide AI by angiotensin-converting enzyme (ACE), and then acts via angiotensin type 1 and type 2 receptors (AT1R and AT2R). Inhibitors of ACE and antagonists of the AT1R are used in the treatment of hypertension, myocardial infarction, and stroke. We now show that the RAAS also plays a major role in autoimmunity, exemplified by multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Using proteomics, we observed that RAAS is up-regulated in brain lesions of MS. AT1R was induced in myelin-specific CD4+ T cells and monocytes during autoimmune neuroinflammation. Blocking AII production with ACE inhibitors or inhibiting AII signaling with AT1R blockers suppressed autoreactive TH1 and TH17 cells and promoted antigen-specific CD4+FoxP3+ regulatory T cells (Treg cells) with inhibition of the canonical NF-kappaB1 transcription factor complex and activation of the alternative NF-kappaB2 pathway. Treatment with ACE inhibitors induces abundant CD4+FoxP3+ T cells with sufficient potency to reverse paralytic EAE. Modulation of the RAAS with inexpensive, safe pharmaceuticals used by millions worldwide is an attractive therapeutic strategy for application to human autoimmune diseases.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Forkhead Transcription Factors/immunology , Humans , Interleukin-17/immunology , Mice , Peptidyl-Dipeptidase A/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/enzymology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/enzymology
19.
Mult Scler J Exp Transl Clin ; 8(3): 20552173221115023, 2022.
Article in English | MEDLINE | ID: mdl-35936922

ABSTRACT

Background: Fingolimod is a sphingosine 1-phosphate receptor modulator approved for relapsing MS. Long-term effects on the immunological profile are not fully understood. Objective: Investigate fingolimod's temporal effects on immune cell subsets, and safety outcomes. Methods: In FLUENT, a 12-month, prospective, non-randomized, open-label, phase IV study, adult participants received fingolimod 0.5 mg/day. Changes in immune cell subsets, anti-John Cunningham virus (JCV) antibody index, and serum neurofilament levels were assessed. Results: 165 fingolimod-naive and 217 participants treated for 2-12 years in routine clinical practice were enrolled. Levels of all monitored peripheral lymphocyte subsets were reduced from month 3 in fingolimod-naive participants. Greatest reductions occurred in naive and central memory CD4+ and CD8+ T cells, and in naive and memory B cells. Most lymphocyte subset levels remained stable in the continuous fingolimod group. Components of the innate immune system remained within reference ranges. No increase in JCV seropositivity was observed. No single cellular subset correlated with anti-JCV antibody index at any time point. Neurofilament levels remained within healthy adult reference limits throughout. No opportunistic infections were reported; no new or unexpected safety signals were observed. Conclusion: FLUENT provides insights into the utility of immunological profiling to evaluate therapy response and potential infection risk.

20.
Mult Scler Relat Disord ; 45: 102347, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32645636

ABSTRACT

Neuromyelitis optica spectrum disorder (NMOSD) is a CNS neuroinflammatory disorder, mediated by the pathogenic autoantibody aquaporin-4 (AQP4-IgG). Current treatment includes long-term use of immunomodulatory therapies, leading to increased rates of infections among this population. It is of interest therefore, to study how the COVID-19 pandemic affects NMOSD patients in terms of their disease activity. A 15-point questionnaire was administered to 33 participants living in Northern California with NMOSD, MS and other related disorders. Although none of the participants were diagnosed with COVID-19, our results show that 2 participants with NMOSD experienced new onset of neurological symptoms and 2 experienced worsening of previous neurological symptoms - suggesting a possible effect of pandemic-related stress on this CNS autoimmune disorder.


Subject(s)
Coronavirus Infections/epidemiology , Coronavirus Infections/psychology , Neuromyelitis Optica/complications , Pneumonia, Viral/epidemiology , Pneumonia, Viral/psychology , Psychological Distress , Symptom Flare Up , Adult , Betacoronavirus , COVID-19 , Female , Humans , Male , Pandemics , Pilot Projects , SARS-CoV-2 , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL