Clinical Focus


  • Multiple Sclerosis
  • Neuroimmunology
  • Clinical Neurophysiology

Academic Appointments


Administrative Appointments


  • Clinical Trials Director, Stanford Multiple Sclerosis and Neuroimmunology Program (2015 - Present)
  • Member, SHC Pharmacy and Therapeutics Specialty Drug Sub-Committee (2016 - 2017)

Honors & Awards


  • Entrance Scholarship, Westminster Hospital Medical School, London, UK (1974)
  • Neuroimmunology Fellow, National Multiple Sclerosis Society (1997-2000)

Boards, Advisory Committees, Professional Organizations


  • Board Member, California Neurological Society (2023 - Present)
  • Member, American Academy of Neurology (1994 - Present)

Professional Education


  • Board Certification: American Board of Psychiatry and Neurology, Neurology (2020)
  • Fellowship: Stanford University Neuroimmunology and Multiple Sclerosis Fellowship (2000) CA
  • Residency: Stanford University Neurology Residency (1997) CA
  • Internship: Stanford University Internal Medicine Residency (1994) CA
  • Medical Education: Westminster Medical School (1980) England

Clinical Trials


  • A Study to Investigate the Safety, Tolerability, and Processing by the Body of Intravenous RO7121932 in Participants With Multiple Sclerosis. Recruiting

    The primary purpose of the study is to evaluate the safety and tolerability of single ascending intravenous (IV) doses of RO7121932 in participants with multiple sclerosis (MS)

    View full details

All Publications


  • Emerging Cerebrospinal Fluid Biomarkers of Disease Activity and Progression in Multiple Sclerosis. JAMA neurology Cross, A. H., Gelfand, J. M., Thebault, S., Bennett, J. L., von Budingen, H. C., Cameron, B., Carruthers, R., Edwards, K., Fallis, R., Gerstein, R., Giacomini, P. S., Greenberg, B., Hafler, D. A., Ionete, C., Kaunzner, U. W., Kodama, L., Lock, C., Longbrake, E. E., Musch, B., Pardo, G., Piehl, F., Weber, M. S., Yuen, S., Ziemssen, T., Bose, G., Freedman, M. S., Anania, V. G., Ramesh, A., Winger, R. C., Jia, X., Herman, A., Harp, C., Bar-Or, A. 2024

    Abstract

    Importance: Biomarkers distinguishing nonrelapsing progressive disease biology from relapsing biology in multiple sclerosis (MS) are lacking. Cerebrospinal fluid (CSF) is an accessible fluid that most closely reflects central nervous system biology.Objective: To identify CSF biological measures associated with progressive MS pathobiology.Design, Setting, and Participants: This cohort study assessed data from 2 prospective MS cohorts: a test cohort provided serial CSF, clinical, and imaging assessments in a multicenter study of patients with relapsing MS (RMS) or primary progressive MS (PPMS) who were initiating anti-CD20 treatment (recruitment: 2016-2018; analysis: 2020-2023). A single-site confirmation cohort was used to assess CSF at baseline and long-term (>10 year) clinical follow-up (analysis: 2022-2023).Exposures: Test-cohort participants initiated standard-of-care ocrelizumab treatment. Confirmation-cohort participants were untreated or received standard-of-care disease-modifying MS therapies.Main Outcomes and Measures: Twenty-five CSF markers, including neurofilament light chain, neurofilament heavy chain, and glial fibrillary acid protein (GFAP); 24-week confirmed disability progression (CDP24); and brain magnetic resonance imaging measures reflecting focal injury, tissue loss, and progressive biology (slowly expanding lesions [SELs]).Results: The test cohort (n=131) included 100 patients with RMS (mean [SD] age, 36.6 [10.4] years; 68 [68%] female and 32 [32%] male; Expanded Disability Status Scale [EDSS] score, 0-5.5), and 31 patients with PPMS (mean [SD] age, 44.9 [7.4] years; 15 [48%] female and 16 [52%] male; EDSS score, 3.0-6.5). The confirmation cohort (n=68) included 41 patients with RMS and 27 with PPMS enrolled at diagnosis (age, 40 years [range, 20-61 years]; 47 [69%] female and 21 [31%] male). In the test cohort, GFAP was correlated with SEL count (r=0.33), greater proportion of T2 lesion volume from SELs (r=0.24), and lower T1-weighted intensity within SELs (r=-0.33) but not with acute inflammatory measures. Neurofilament heavy chain was correlated with SEL count (r=0.25) and lower T1-weighted intensity within SELs (r=-0.28). Immune markers correlated with measures of acute inflammation and, unlike GFAP, were impacted by anti-CD20. In the confirmation cohort, higher baseline CSF GFAP levels were associated with long-term CDP24 (hazard ratio, 2.1; 95% CI, 1.3-3.4; P=.002).Conclusions and Relevance: In this study, activated glial markers (in particular GFAP) and neurofilament heavy chain were associated specifically with nonrelapsing progressive disease outcomes (independent of acute inflammatory activity). Elevated CSF GFAP was associated with long-term MS disease progression.

    View details for DOI 10.1001/jamaneurol.2024.0017

    View details for PubMedID 38466277

  • Neural Network Corrections to Intermolecular Interaction Terms of a Molecular Force Field Capture Nuclear Quantum Effects in Calculations of Liquid Thermodynamic Properties. Journal of chemical theory and computation Kurnikov, I. V., Pereyaslavets, L., Kamath, G., Sakipov, S. N., Voronina, E., Butin, O., Illarionov, A., Leontyev, I., Nawrocki, G., Darkhovskiy, M., Olevanov, M., Ivahnenko, I., Chen, Y., Lock, C. B., Levitt, M., Kornberg, R. D., Fain, B. 2024

    Abstract

    We incorporate nuclear quantum effects (NQE) in condensed matter simulations by introducing short-range neural network (NN) corrections to the ab initio fitted molecular force field ARROW. Force field NN corrections are fitted to average interaction energies and forces of molecular dimers, which are simulated using the Path Integral Molecular Dynamics (PIMD) technique with restrained centroid positions. The NN-corrected force field allows reproduction of the NQE for computed liquid water and methane properties such as density, radial distribution function (RDF), heat of evaporation (HVAP), and solvation free energy. Accounting for NQE through molecular force field corrections circumvents the need for explicit computationally expensive PIMD simulations in accurate calculations of the properties of chemical and biological systems. The accuracy and locality of pairwise NN NQE corrections indicate that this approach could be applicable to complex heterogeneous systems, such as proteins.

    View details for DOI 10.1021/acs.jctc.3c00921

    View details for PubMedID 38240485

  • Combining Force Fields and Neural Networks for an Accurate Representation of Bonded Interactions. The journal of physical chemistry. A Kamath, G., Illarionov, A., Sakipov, S., Pereyaslavets, L., Kurnikov, I. V., Butin, O., Voronina, E., Ivahnenko, I., Leontyev, I., Nawrocki, G., Darkhovskiy, M., Olevanov, M., Cherniavskyi, Y. K., Lock, C., Greenslade, S., Chen, Y., Kornberg, R. D., Levitt, M., Fain, B. 2024

    Abstract

    We present a formalism of a neural network encoding bonded interactions in molecules. This intramolecular encoding is consistent with the models of intermolecular interactions previously designed by this group. Variants of the encoding fed into a corresponding neural network may be used to economically improve the representation of torsional degrees of freedom in any force field. We test the accuracy of the reproduction of the ab initio potential energy surface on a set of conformations of two dipeptides, methyl-capped ALA and ASP, in several scenarios. The encoding, either alone or in conjunction with an analytical potential, improves agreement with ab initio energies that are on par with those of other neural network-based potentials. Using the encoding and neural nets in tandem with an analytical model places the agreements firmly within "chemical accuracy" of ±0.5 kcal/mol.

    View details for DOI 10.1021/acs.jpca.3c07598

    View details for PubMedID 38232765

  • Prevalence, Demographic, and Clinical Factors Associated With Cognitive Dysfunction in Patients With Neuromyelitis Optica Spectrum Disorder. Neurology Vlahovic, L., McDonald, J., Hinman, J., Tomczak, A., Lock, C., Palmer, C. A., Cook, L. J., Yeaman, M. R., Burnett, M. K., Deutsch, G. K., Nelson, L. M., Han, M. H. 2024; 102 (1): e207965

    Abstract

    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.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.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.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.

    View details for DOI 10.1212/WNL.0000000000207965

    View details for PubMedID 38165361

  • Combining Force Fields and Neural Networks for an Accurate Representation of Chemically Diverse Molecular Interactions. Journal of the American Chemical Society Illarionov, A., Sakipov, S., Pereyaslavets, L., Kurnikov, I. V., Kamath, G., Butin, O., Voronina, E., Ivahnenko, I., Leontyev, I., Nawrocki, G., Darkhovskiy, M., Olevanov, M., Cherniavskyi, Y. K., Lock, C., Greenslade, S., Sankaranarayanan, S. K., Kurnikova, M. G., Potoff, J., Kornberg, R. D., Levitt, M., Fain, B. 2023

    Abstract

    A key goal of molecular modeling is the accurate reproduction of the true quantum mechanical potential energy of arbitrary molecular ensembles with a tractable classical approximation. The challenges are that analytical expressions found in general purpose force fields struggle to faithfully represent the intermolecular quantum potential energy surface at close distances and in strong interaction regimes; that the more accurate neural network approximations do not capture crucial physics concepts, e.g., nonadditive inductive contributions and application of electric fields; and that the ultra-accurate narrowly targeted models have difficulty generalizing to the entire chemical space. We therefore designed a hybrid wide-coverage intermolecular interaction model consisting of an analytically polarizable force field combined with a short-range neural network correction for the total intermolecular interaction energy. Here, we describe the methodology and apply the model to accurately determine the properties of water, the free energy of solvation of neutral and charged molecules, and the binding free energy of ligands to proteins. The correction is subtyped for distinct chemical species to match the underlying force field, to segment and reduce the amount of quantum training data, and to increase accuracy and computational speed. For the systems considered, the hybrid ab initio parametrized Hamiltonian reproduces the two-body dimer quantum mechanics (QM) energies to within 0.03 kcal/mol and the nonadditive many-molecule contributions to within 2%. Simulations of molecular systems using this interaction model run at speeds of several nanoseconds per day.

    View details for DOI 10.1021/jacs.3c07628

    View details for PubMedID 37856313

  • SARS-CoV-2 Vaccine Immune Response on Anti-Complement Therapy, Eculizumab Sattarnezhad, N., Sumera, J., McDonald, J., Nie, E., Tomczak, A., Joseph, Y., Kalle, S., Sarkar, T., Kipp, L., Lock, C., Dunn, J. E., Han, M. LIPPINCOTT WILLIAMS & WILKINS. 2023
  • Progression of Mycosis Fungoides After Fingolimod Treatment for Multiple Sclerosis and Targeted Next-Generation Sequencing Demonstrating Potential Links Between the Two Diseases. JCO precision oncology Narala, S., Che, Y., Saleem, A., Lock, C. B., Kim, Y. H., Rieger, K. E. 2023; 7: e2200501

    View details for DOI 10.1200/PO.22.00501

    View details for PubMedID 36724412

  • Differential effects of anti-CD20 therapy on CD4 and CD8 T cells and implication of CD20-expressing CD8 T cells in MS disease activity. Proceedings of the National Academy of Sciences of the United States of America Shinoda, K., Li, R., Rezk, A., Mexhitaj, I., Patterson, K. R., Kakara, M., Zuroff, L., Bennett, J. L., von Büdingen, H. C., Carruthers, R., Edwards, K. R., Fallis, R., Giacomini, P. S., Greenberg, B. M., Hafler, D. A., Ionete, C., Kaunzner, U. W., Lock, C. B., Longbrake, E. E., Pardo, G., Piehl, F., Weber, M. S., Ziemssen, T., Jacobs, D., Gelfand, J. M., Cross, A. H., Cameron, B., Musch, B., Winger, R. C., Jia, X., Harp, C. T., Herman, A., Bar-Or, A. 2023; 120 (3): e2207291120

    Abstract

    A small proportion of multiple sclerosis (MS) patients develop new disease activity soon after starting anti-CD20 therapy. This activity does not recur with further dosing, possibly reflecting deeper depletion of CD20-expressing cells with repeat infusions. We assessed cellular immune profiles and their association with transient disease activity following anti-CD20 initiation as a window into relapsing disease biology. Peripheral blood mononuclear cells from independent discovery and validation cohorts of MS patients initiating ocrelizumab were assessed for phenotypic and functional profiles using multiparametric flow cytometry. Pretreatment CD20-expressing T cells, especially CD20dimCD8+ T cells with a highly inflammatory and central nervous system (CNS)-homing phenotype, were significantly inversely correlated with pretreatment MRI gadolinium-lesion counts, and also predictive of early disease activity observed after anti-CD20 initiation. Direct removal of pretreatment proinflammatory CD20dimCD8+ T cells had a greater contribution to treatment-associated changes in the CD8+ T cell pool than was the case for CD4+ T cells. Early disease activity following anti-CD20 initiation was not associated with reconstituting CD20dimCD8+ T cells, which were less proinflammatory compared with pretreatment. Similarly, this disease activity did not correlate with early reconstituting B cells, which were predominantly transitional CD19+CD24highCD38high with a more anti-inflammatory profile. We provide insights into the mode-of-action of anti-CD20 and highlight a potential role for CD20dimCD8+ T cells in MS relapse biology; their strong inverse correlation with both pretreatment and early posttreatment disease activity suggests that CD20-expressing CD8+ T cells leaving the circulation (possibly to the CNS) play a particularly early role in the immune cascades involved in relapse development.

    View details for DOI 10.1073/pnas.2207291120

    View details for PubMedID 36634138

  • Protein-Ligand Binding Free-Energy Calculations with ARROW─A Purely First-Principles Parameterized Polarizable Force Field. Journal of chemical theory and computation Nawrocki, G., Leontyev, I., Sakipov, S., Darkhovskiy, M., Kurnikov, I., Pereyaslavets, L., Kamath, G., Voronina, E., Butin, O., Illarionov, A., Olevanov, M., Kostikov, A., Ivahnenko, I., Patel, D. S., Sankaranarayanan, S. K., Kurnikova, M. G., Lock, C., Crooks, G. E., Levitt, M., Kornberg, R. D., Fain, B. 2022

    Abstract

    Protein-ligand binding free-energy calculations using molecular dynamics (MD) simulations have emerged as a powerful tool for in silico drug design. Here, we present results obtained with the ARROW force field (FF)─a multipolar polarizable and physics-based model with all parameters fitted entirely to high-level ab initio quantum mechanical (QM) calculations. ARROW has already proven its ability to determine solvation free energy of arbitrary neutral compounds with unprecedented accuracy. The ARROW FF parameterization is now extended to include coverage of all amino acids including charged groups, allowing molecular simulations of a series of protein-ligand systems and prediction of their relative binding free energies. We ensure adequate sampling by applying a novel technique that is based on coupling the Hamiltonian Replica exchange (HREX) with a conformation reservoir generated via potential softening and nonequilibrium MD. ARROW provides predictions with near chemical accuracy (mean absolute error of 0.5 kcal/mol) for two of the three protein systems studied here (MCL1 and Thrombin). The third protein system (CDK2) reveals the difficulty in accurately describing dimer interaction energies involving polar and charged species. Overall, for all of the three protein systems studied here, ARROW FF predicts relative binding free energies of ligands with a similar accuracy level as leading nonpolarizable force fields.

    View details for DOI 10.1021/acs.jctc.2c00930

    View details for PubMedID 36459593

  • Evaluation of neurotrophic factor secreting mesenchymal stem cells in progressive multiple sclerosis MULTIPLE SCLEROSIS JOURNAL Cohen, J. A., Lublin, F. D., Lock, C., Pelletier, D., Chitnis, T., Mehra, M., Gothelf, Y., Aricha, R., Lindborg, S., Lebovits, C., Levy, Y., Motamed Khorasani, A., Kern, R. 2022: 13524585221122156

    Abstract

    Autologous mesenchymal stem cell neurotrophic factor-secreting cells (NurOwn®) have the potential to modify underlying disease mechanisms in progressive multiple sclerosis (PMS).This open-label phase II study was conducted to evaluate safety/efficacy of three intrathecal cell treatments.Eighteen participants with non-relapsing PMS were treated. The primary endpoint was safety. Secondary endpoints included: cerebrospinal fluid (CSF) biomarkers; timed 25-foot walk speed, nine-hole peg test (9-HPT), low-contrast letter acuity, symbol digit modalities test, and 12-item multiple sclerosis (MS) walking scale. Seventeen participants received all treatments.No deaths/adverse events related to worsening of MS, clinical/magnetic resonance imaging (MRI) evidence of disease activation, and clinically significant changes in safety lab results were reported. Two participants developed symptoms of low back and leg pain, consistent with a diagnosis of arachnoiditis, occurring in one of three intrathecal treatments in both participants. Nineteen percent of treated participants achieved pre-specified ⩾ 25% improvements in timed 25-foot walk speed/nine-HPT at 28 weeks compared to baseline, along with consistent efficacy signals for pre-specified response criteria across other secondary efficacy outcomes. CSF neuroprotective factors increased, and inflammatory biomarkers decreased after treatment, consistent with the proposed mechanism of action.Based on these encouraging preliminary findings, further confirmation in a randomized study is warranted.

    View details for DOI 10.1177/13524585221122156

    View details for Web of Science ID 000853962400001

    View details for PubMedID 36113170

  • Clonally Expanded B Cells in Multiple Sclerosis Bind EBV EBNA1 and GlialCAM. Nature Lanz, T. V., Brewer, R. C., Ho, P. P., Moon, J. S., Jude, K. M., Fernandez, D., Fernandes, R. A., Gomez, A. M., Nadj, G. S., Bartley, C. M., Schubert, R. D., Hawes, I. A., Vazquez, S. E., Iyer, M., Zuchero, J. B., Teegen, B., Dunn, J. E., Lock, C. B., Kipp, L. B., Cotham, V. C., Ueberheide, B. M., Aftab, B. T., Anderson, M. S., DeRisi, J. L., Wilson, M. R., Bashford-Rogers, R. J., Platten, M., Garcia, K. C., Steinman, L., Robinson, W. H. 2022

    Abstract

    Multiple sclerosis (MS) is a heterogenous autoimmune disease in which autoreactive lymphocytes attack the myelin sheath of the central nervous system (CNS). B lymphocytes in the cerebrospinal fluid (CSF) of MS patients contribute to inflammation and secrete oligoclonal immunoglobulins1,2. Epstein-Barr virus (EBV) infection has been linked to MS epidemiologically, but its pathological role remains unclear3. Here we demonstrate high-affinity molecular mimicry between the EBV transcription factor EBNA1 and the CNS protein GlialCAM, and provide structural and in-vivo functional evidence for its relevance. A cross-reactive CSF-derived antibody was initially identified by single-cell sequencing of the paired-chain B cell repertoire of MS blood and CSF, followed by protein microarray-based testing of recombinantly expressed CSF-derived antibodies against MS-associated viruses. Sequence analysis, affinity measurements, and the crystal structure of the EBNA1-peptide epitope in complex with the autoreactive Fab fragment allowed for tracking the development of the naïve EBNA1-restricted antibody to a mature EBNA1/GlialCAM cross-reactive antibody. Molecular mimicry is facilitated by a post-translational modification of GlialCAM. EBNA1 immunization exacerbates the mouse model of MS and anti-EBNA1/GlialCAM antibodies are prevalent in MS patients. Our results provide a mechanistic link for the association between MS and EBV, and could guide the development of novel MS therapies.

    View details for DOI 10.1038/s41586-022-04432-7

    View details for PubMedID 35073561

  • MR susceptibility contrast imaging using a 2D simultaneous multi-slice gradient-echo sequence at 7T. PloS one Bian, W., Kerr, A. B., Tranvinh, E., Parivash, S., Zahneisen, B., Han, M. H., Lock, C. B., Goubran, M., Zhu, K., Rutt, B. K., Zeineh, M. M. 2019; 14 (7): e0219705

    Abstract

    PURPOSE: To develop a 7T simultaneous multi-slice (SMS) 2D gradient-echo sequence for susceptibility contrast imaging, and to compare its quality to 3D imaging.METHODS: A frequency modulated and phase cycled RF pulse was designed to simultaneously excite multiple slices in multi-echo 2D gradient-echo imaging. The imaging parameters were chosen to generate images with susceptibility contrast, including T2*-weighted magnitude/phase images, susceptibility-weighted images and quantitative susceptibility/R2* maps. To compare their image quality with 3D gradient-echo imaging, both 2D and 3D imaging were performed on 11 healthy volunteers and 4 patients with multiple sclerosis (MS). The signal to noise ratio (SNR) in gray and white matter and their contrast to noise ratio (CNR) was simulated for the 2D and 3D magnitude images using parameters from the imaging. The experimental SNRs and CNRs were measured in gray/white matter and deep gray matter structures on magnitude, phase, R2* and QSM images from volunteers and the visibility of MS lesions on these images from patients was visually rated. All SNRs and CNRs were compared between the 2D and 3D imaging using a paired t-test.RESULTS: Although the 3D magnitude images still had significantly higher SNRs (by 13.0~17.6%), the 2D magnitude and QSM images generated significantly higher gray/white matter or globus pallidus/putamen contrast (by 13.3~87.5%) and significantly higher MS lesion contrast (by 5.9~17.3%).CONCLUSION: 2D SMS gradient-echo imaging can serve as an alternative to often used 3D imaging to obtain susceptibility-contrast-weighted images, with an advantage of providing better image contrast and MS lesion sensitivity.

    View details for DOI 10.1371/journal.pone.0219705

    View details for PubMedID 31314813

  • Autoimmune Hepatitis During Treatment of Multiple Sclerosis with Alemtuzumab Carlson, A., Bozinov, N., Kipp, L., Dunn, J., Lock, C. LIPPINCOTT WILLIAMS & WILKINS. 2018
  • A Multi-scale Multiple Sclerosis Lesion Change Detection in a Multi-sequence MRI Cheng, M., Galimzianova, A., Lesjak, Z., Spiclin, Z., Lock, C. B., Rubin, D. L., Stoyanov, D., Taylor, Z., Carneiro, G., SyedaMahmood, T. SPRINGER INTERNATIONAL PUBLISHING AG. 2018: 353–60
  • Janus-like opposing roles of CD47 in autoimmune brain inflammation in humans and mice JOURNAL OF EXPERIMENTAL MEDICINE Han, M. H., Lundgren, D. H., Jaiswal, S., Chao, M., Graham, K. L., Garris, C. S., Axtell, R. C., Ho, P. P., Lock, C. B., Woodard, J. I., Brownell, S. E., Zoudilova, M., Hunt, J. F., Baranzini, S. E., Butcher, E. C., Raine, C. S., Sobel, R. A., Han, D. K., Weissman, I., Steinman, L. 2012; 209 (7): 1325-1334

    Abstract

    Comparison of transcriptomic and proteomic data from pathologically similar multiple sclerosis (MS) lesions reveals down-regulation of CD47 at the messenger RNA level and low abundance at the protein level. Immunohistochemical studies demonstrate that CD47 is expressed in normal myelin and in foamy macrophages and reactive astrocytes within active MS lesions. We demonstrate that CD47(-/-) mice are refractory to experimental autoimmune encephalomyelitis (EAE), primarily as the result of failure of immune cell activation after immunization with myelin antigen. In contrast, blocking with a monoclonal antibody against CD47 in mice at the peak of paralysis worsens EAE severity and enhances immune activation in the peripheral immune system. In vitro assays demonstrate that blocking CD47 also promotes phagocytosis of myelin and that this effect is dependent on signal regulatory protein α (SIRP-α). Immune regulation and phagocytosis are mechanisms for CD47 signaling in autoimmune neuroinflammation. Depending on the cell type, location, and disease stage, CD47 has Janus-like roles, with opposing effects on EAE pathogenesis.

    View details for DOI 10.1084/jem.20101974

    View details for Web of Science ID 000306174300008

    View details for PubMedID 22734047

    View details for PubMedCentralID PMC3405500

  • Inhibitory role for GABA in autoimmune inflammation PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Bhat, R., Axtell, R., Mitra, A., Miranda, M., Lock, C., Tsien, R. W., Steinman, L. 2010; 107 (6): 2580-2585

    Abstract

    GABA, the principal inhibitory neurotransmitter in the adult brain, has a parallel inhibitory role in the immune system. We demonstrate that immune cells synthesize GABA and have the machinery for GABA catabolism. Antigen-presenting cells (APCs) express functional GABA receptors and respond electrophysiologically to GABA. Thus, the immune system harbors all of the necessary constituents for GABA signaling, and GABA itself may function as a paracrine or autocrine factor. These observations led us to ask further whether manipulation of the GABA pathway influences an animal model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Increasing GABAergic activity ameliorates ongoing paralysis in EAE via inhibition of inflammation. GABAergic agents act directly on APCs, decreasing MAPK signals and diminishing subsequent adaptive inflammatory responses to myelin proteins.

    View details for DOI 10.1073/pnas.0915139107

    View details for Web of Science ID 000274408100041

    View details for PubMedID 20133656

    View details for PubMedCentralID PMC2823917

  • Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis NATURE MEDICINE Lock, C., Hermans, G., Pedotti, R., Brendolan, A., Schadt, E., Garren, H., Langer-Gould, A., Strober, S., Cannella, B., Allard, J., Klonowski, P., Austin, A., Lad, N., Kaminski, N., GALLI, S. J., Oksenberg, J. R., Raine, C. S., Heller, R., Steinman, L. 2002; 8 (5): 500-508

    Abstract

    Microarray analysis of multiple sclerosis (MS) lesions obtained at autopsy revealed increased transcripts of genes encoding inflammatory cytokines, particularly interleukin-6 and -17, interferon-gamma and associated downstream pathways. Comparison of two poles of MS pathology--acute lesions with inflammation versus 'silent' lesions without inflammation--revealed differentially transcribed genes. Some products of these genes were chosen as targets for therapy of experimental autoimmune encephalomyelitis (EAE) in mice. Granulocyte colony-stimulating factor is upregulated in acute, but not in chronic, MS lesions, and the effect on ameliorating EAE is more pronounced in the acute phase, in contrast to knocking out the immunoglobulin Fc receptor common gamma chain where the effect is greatest on chronic disease. These results in EAE corroborate the microarray studies on MS lesions. Large-scale analysis of transcripts in MS lesions elucidates new aspects of pathology and opens possibilities for therapy.

    View details for DOI 10.1038/nm0502-500

    View details for PubMedID 11984595

  • The influence of the proinflammatory cytokine, osteopontin, on autoimmune demyelinating disease SCIENCE Chabas, D., Baranzini, S. E., Mitchell, D., Bernard, C. C., Rittling, S. R., Denhardt, D. T., Sobel, R. A., Lock, C., Karpuj, M., Pedotti, R., Heller, R., Oksenberg, J. R., Steinman, L. 2001; 294 (5547): 1731-1735

    Abstract

    Multiple sclerosis is a demyelinating disease, characterized by inflammation in the brain and spinal cord, possibly due to autoimmunity. Large-scale sequencing of cDNA libraries, derived from plaques dissected from brains of patients with multiple sclerosis (MS), indicated an abundance of transcripts for osteopontin (OPN). Microarray analysis of spinal cords from rats paralyzed by experimental autoimmune encephalomyelitis (EAE), a model of MS, also revealed increased OPN transcripts. Osteopontin-deficient mice were resistant to progressive EAE and had frequent remissions, and myelin-reactive T cells in OPN-/- mice produced more interleukin 10 and less interferon-gamma than in OPN+/+ mice. Osteopontin thus appears to regulate T helper cell-1 (TH1)-mediated demyelinating disease, and it may offer a potential target in blocking development of progressive MS.

    View details for Web of Science ID 000172307400049

    View details for PubMedID 11721059

  • The role of TNF alpha and lymphotoxin in demyelinating disease ANNALS OF THE RHEUMATIC DISEASES Lock, C., Oksenberg, J., Steinman, L. 1999; 58: 121-128
  • The role of TNFalpha and lymphotoxin in demyelinating disease. Annals of the rheumatic diseases Lock, C., Oksenberg, J., Steinman, L. 1999; 58: I121-8

    View details for PubMedID 10577988

  • EXPRESSION OF HLA-DR4 AND HUMAN CD4 TRANSGENES IN MICE DETERMINES THE VARIABLE REGION BETA-CHAIN T-CELL REPERTOIRE AND MEDIATES AN HLA-DR-RESTRICTED IMMUNE-RESPONSE PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Fugger, L., Michie, S. A., Rulifson, I., Lock, C. B., McDevitt, G. S. 1994; 91 (13): 6151-6155

    Abstract

    Inherited susceptibility to rheumatoid arthritis is associated with genes encoding the human major histocompatibility complex class II molecule HLA-DR4. To study the immune function of HLA-DR4 and attempt to generate a murine model of rheumatoid arthritis we have produced triple transgenic mice expressing HLA-DRA*0101, -DRB1*0401, and human CD4. The expression of the HLA transgenes is driven by the promoter of the murine major histocompatibility complex class II I-E alpha gene and was found on murine cells that normally display major histocompatibility complex class II molecules. The expression of the human CD4 transgene is driven by the murine CD3 delta-promoter, and therefore its gene product was found on cells that express murine CD3. In contrast to other HLA-DR and HLA-DQ transgenic mouse lines, the transgenes are functional in our mice. In H-2 I-E-negative transgenic mice, T cells expressing variable region beta chain (V beta) 3, 5, 6, 7, 9, 11, 12, or 13 were either absent or significantly reduced, in contrast to H-2 I-E-negative nontransgenic littermates. In addition, the peptide antigen influenza A virus hemagglutinin 307-319, which binds to the HLA-DRA*0101/-DRB1*0401 heterodimer with high affinity and induces an HLA-DR-restricted and CD4+ T-cell response in humans, also induced a T-cell response in the triple transgenic mice but not in nontransgenic littermates. Thus, these transgenic mice should permit extensive testing of the antigen-presentation capabilities of the HLA-DRA*0101/-DRB1*0401 molecule.

    View details for Web of Science ID A1994NT46100086

    View details for PubMedID 8016129

  • MINIMUM STRUCTURAL REQUIREMENTS FOR PEPTIDE PRESENTATION BY MAJOR HISTOCOMPATIBILITY COMPLEX CLASS-II MOLECULES - IMPLICATIONS IN INDUCTION OF AUTOIMMUNITY PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Gautam, A. M., Lock, C. B., Smilek, D. E., Pearson, C. I., Steinman, L., McDevitt, H. O. 1994; 91 (2): 767-771

    Abstract

    The precise mechanisms of failure of immunological tolerance to self proteins are not known. Major histocompatibility complex (MHC) susceptibility alleles, the target peptides, and T cells with anti-self reactivity must be present to cause autoimmune diseases. Experimental autoimmune encephalomyelitis (EAE) is a murine model of a human autoimmune disease, multiple sclerosis. In EAE, residues 1-11 of myelin basic protein (MBP) are the dominant disease-inducing determinants in PL/J and (PL/J x SJL/J)F1 mice. Here we report that a six-residue peptide (five of them native) of MBP can induce EAE. Using peptide analogues of the MBP-(1-11) peptide, we demonstrate that only four native MBP residues are required to stimulate MBP-specific T cells. Therefore, this study demonstrates lower minimum structural requirements for effective antigen presentation by MHC class II molecules. Many viral and bacterial proteins share short runs of amino acid similarity with host self proteins, a phenomenon known as molecular mimicry. Since a six-residue peptide can sensitize MBP-specific T cells to cause EAE, these results define a minimum sequence identity for molecular mimicry in autoimmunity.

    View details for Web of Science ID A1994MR98900070

    View details for PubMedID 7507253

  • ANTIGEN RECOGNITION AND PEPTIDE-MEDIATED IMMUNOTHERAPY IN AUTOIMMUNE-DISEASE IMMUNOLOGICAL REVIEWS Smilek, D. E., Lock, C. B., McDevitt, H. O. 1990; 118: 37-71

    View details for Web of Science ID A1990EU70000003

    View details for PubMedID 1706681