1
|
Falk I, Maric D, Leibovitch E, Sati P, Lefeuvre J, Luciano NJ, Guy J, Ha SK, Owen DR, Aigbirhio F, Matthews PM, Reich DS, Jacobson S. Characteristics of TSPO expression in marmoset EAE. J Neuroinflammation 2025; 22:19. [PMID: 39871344 PMCID: PMC11773908 DOI: 10.1186/s12974-025-03343-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/10/2025] [Indexed: 01/29/2025] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) and is a leading non-traumatic cause of disability in young adults. The 18 kDa Translocator Protein (TSPO) is a mitochondrial protein and positron emission tomography (PET)-imaging target that is highly expressed in MS brain lesions. It is used as an inflammatory biomarker and has been proposed as a therapeutic target. However, its specific pathological significance in humans is not well understood. Experimental autoimmune encephalomyelitis (EAE) in the common marmoset is a well-established primate model of MS. Studying TSPO expression in this model will enhance our understanding of its expression in MS. This study therefore characterizes patterns of TSPO expression in fixed CNS tissues from one non-EAE control marmoset and 8 EAE marmosets using multiplex immunofluorescence. In control CNS tissue, we find that TSPO is expressed in the leptomeninges, ependyma, and over two-thirds of Iba1 + microglia, but not astrocytes or neurons. In Iba1 + cells in both control and acute EAE tissue, we find that TSPO is co-expressed with markers of antigen presentation (CD74), early activation (MRP14), phagocytosis (CD163) and anti-inflammatory phenotype (Arg1); a high level of TSPO expression is not restricted to a particular microglial phenotype. While TSPO is expressed in over 88% of activated Iba1 + cells in acute lesions in marmoset EAE, it also is sometimes observed in subsets of astrocytes and neurons. Additionally, we find the percentage of Iba1 + cells expressing TSPO declines significantly in lesions > 5 months old and may be as low as 13% in chronic lesions. However, we also find increased astrocytic TSPO expression in chronic-appearing lesions with astrogliosis. Finally, we find expression of TSPO in a subset of neurons, most frequently GLS2 + glutamatergic neurons. The shift in TSPO expression from Iba + microglia/macrophages to astrocytes over time is similar to patterns suggested by earlier neuropathology studies in MS. Thus, marmoset EAE appears to be a clinically relevant model for the study of TSPO in immune dysregulation in human disease.
Collapse
Affiliation(s)
- Irene Falk
- Viral Immunology Section, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Building 10, Room 5C103, 10 Center Drive, Bethesda, MD, 20892-1400, USA
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Emily Leibovitch
- Viral Immunology Section, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Building 10, Room 5C103, 10 Center Drive, Bethesda, MD, 20892-1400, USA
| | - Pascal Sati
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Lefeuvre
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas J Luciano
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Joseph Guy
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Seung-Kwon Ha
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David R Owen
- Department of Brain Sciences, Imperial College London, London, UK
| | - Franklin Aigbirhio
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Steven Jacobson
- Viral Immunology Section, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Building 10, Room 5C103, 10 Center Drive, Bethesda, MD, 20892-1400, USA.
| |
Collapse
|
2
|
Oertel FC, Hastermann M, Paul F. Delimiting MOGAD as a disease entity using translational imaging. Front Neurol 2023; 14:1216477. [PMID: 38333186 PMCID: PMC10851159 DOI: 10.3389/fneur.2023.1216477] [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: 05/03/2023] [Accepted: 08/23/2023] [Indexed: 02/10/2024] Open
Abstract
The first formal consensus diagnostic criteria for myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) were recently proposed. Yet, the distinction of MOGAD-defining characteristics from characteristics of its important differential diagnoses such as multiple sclerosis (MS) and aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorder (NMOSD) is still obstructed. In preclinical research, MOG antibody-based animal models were used for decades to derive knowledge about MS. In clinical research, people with MOGAD have been combined into cohorts with other diagnoses. Thus, it remains unclear to which extent the generated knowledge is specifically applicable to MOGAD. Translational research can contribute to identifying MOGAD characteristic features by establishing imaging methods and outcome parameters on proven pathophysiological grounds. This article reviews suitable animal models for translational MOGAD research and the current state and prospect of translational imaging in MOGAD.
Collapse
Affiliation(s)
- Frederike Cosima Oertel
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Hastermann
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
3
|
Nutma E, Fancy N, Weinert M, Tsartsalis S, Marzin MC, Muirhead RCJ, Falk I, Breur M, de Bruin J, Hollaus D, Pieterman R, Anink J, Story D, Chandran S, Tang J, Trolese MC, Saito T, Saido TC, Wiltshire KH, Beltran-Lobo P, Phillips A, Antel J, Healy L, Dorion MF, Galloway DA, Benoit RY, Amossé Q, Ceyzériat K, Badina AM, Kövari E, Bendotti C, Aronica E, Radulescu CI, Wong JH, Barron AM, Smith AM, Barnes SJ, Hampton DW, van der Valk P, Jacobson S, Howell OW, Baker D, Kipp M, Kaddatz H, Tournier BB, Millet P, Matthews PM, Moore CS, Amor S, Owen DR. Translocator protein is a marker of activated microglia in rodent models but not human neurodegenerative diseases. Nat Commun 2023; 14:5247. [PMID: 37640701 PMCID: PMC10462763 DOI: 10.1038/s41467-023-40937-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
Microglial activation plays central roles in neuroinflammatory and neurodegenerative diseases. Positron emission tomography (PET) targeting 18 kDa Translocator Protein (TSPO) is widely used for localising inflammation in vivo, but its quantitative interpretation remains uncertain. We show that TSPO expression increases in activated microglia in mouse brain disease models but does not change in a non-human primate disease model or in common neurodegenerative and neuroinflammatory human diseases. We describe genetic divergence in the TSPO gene promoter, consistent with the hypothesis that the increase in TSPO expression in activated myeloid cells depends on the transcription factor AP1 and is unique to a subset of rodent species within the Muroidea superfamily. Finally, we identify LCP2 and TFEC as potential markers of microglial activation in humans. These data emphasise that TSPO expression in human myeloid cells is related to different phenomena than in mice, and that TSPO-PET signals in humans reflect the density of inflammatory cells rather than activation state.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
- Department of Neurobiology and Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Nurun Fancy
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Maria Weinert
- Department of Brain Sciences, Imperial College London, London, UK
| | - Stergios Tsartsalis
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Manuel C Marzin
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Robert C J Muirhead
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Irene Falk
- Viral Immunology Section, NIH, Bethesda, MD, USA
- Flow and Imaging Cytometry Core Facility, NIH, Bethesda, MD, USA
| | - Marjolein Breur
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Joy de Bruin
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Robin Pieterman
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Jasper Anink
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - David Story
- UK Dementia Research Institute at Edinburgh, Edinburgh, UK
| | | | - Jiabin Tang
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Maria C Trolese
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama, Japan
| | - Takaomi C Saido
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Nagoya, Japan
| | | | - Paula Beltran-Lobo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alexandra Phillips
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Jack Antel
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Luke Healy
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Marie-France Dorion
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Dylan A Galloway
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Rochelle Y Benoit
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Quentin Amossé
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Kelly Ceyzériat
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | | | - Enikö Kövari
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Caterina Bendotti
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Carola I Radulescu
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Jia Hui Wong
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Anna M Barron
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Amy M Smith
- UK Dementia Research Institute at Imperial College London, London, UK
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Samuel J Barnes
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | | | - Paul van der Valk
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | | | - Owain W Howell
- Institute of Life Science (ILS), Swansea University Medical School, Swansea, UK
| | - David Baker
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany
| | - Hannes Kaddatz
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany
| | | | - Philippe Millet
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
- Division of Adult Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Craig S Moore
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands.
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK.
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany.
| | - David R Owen
- Department of Brain Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute at Imperial College London, London, UK.
| |
Collapse
|
4
|
Yang HJ, Wang D, Wen X, Weiner DM, Via LE. One Size Fits All? Not in In Vivo Modeling of Tuberculosis Chemotherapeutics. Front Cell Infect Microbiol 2021; 11:613149. [PMID: 33796474 PMCID: PMC8008060 DOI: 10.3389/fcimb.2021.613149] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Tuberculosis (TB) remains a global health problem despite almost universal efforts to provide patients with highly effective chemotherapy, in part, because many infected individuals are not diagnosed and treated, others do not complete treatment, and a small proportion harbor Mycobacterium tuberculosis (Mtb) strains that have become resistant to drugs in the standard regimen. Development and approval of new drugs for TB have accelerated in the last 10 years, but more drugs are needed due to both Mtb's development of resistance and the desire to shorten therapy to 4 months or less. The drug development process needs predictive animal models that recapitulate the complex pathology and bacterial burden distribution of human disease. The human host response to pulmonary infection with Mtb is granulomatous inflammation usually resulting in contained lesions and limited bacterial replication. In those who develop progressive or active disease, regions of necrosis and cavitation can develop leading to lasting lung damage and possible death. This review describes the major vertebrate animal models used in evaluating compound activity against Mtb and the disease presentation that develops. Each of the models, including the zebrafish, various mice, guinea pigs, rabbits, and non-human primates provides data on number of Mtb bacteria and pathology resolution. The models where individual lesions can be dissected from the tissue or sampled can also provide data on lesion-specific bacterial loads and lesion-specific drug concentrations. With the inclusion of medical imaging, a compound's effect on resolution of pathology within individual lesions and animals can also be determined over time. Incorporation of measurement of drug exposure and drug distribution within animals and their tissues is important for choosing the best compounds to push toward the clinic and to the development of better regimens. We review the practical aspects of each model and the advantages and limitations of each in order to promote choosing a rational combination of them for a compound's development.
Collapse
Affiliation(s)
- Hee-Jeong Yang
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Decheng Wang
- Medical College, China Three Gorges University, Yichang, China.,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
| | - Xin Wen
- Medical College, China Three Gorges University, Yichang, China.,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
| | - Danielle M Weiner
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States.,Tuberculosis Imaging Program, DIR, NIAID, NIH, Bethesda, MD, United States
| | - Laura E Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States.,Tuberculosis Imaging Program, DIR, NIAID, NIH, Bethesda, MD, United States.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
5
|
Chase Huizar C, Raphael I, Forsthuber TG. Genomic, proteomic, and systems biology approaches in biomarker discovery for multiple sclerosis. Cell Immunol 2020; 358:104219. [PMID: 33039896 PMCID: PMC7927152 DOI: 10.1016/j.cellimm.2020.104219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is a neuroinflammatory disorder characterized by autoimmune-mediated inflammatory lesions in CNS leading to myelin damage and axonal loss. MS is a heterogenous disease with variable and unpredictable disease course. Due to its complex nature, MS is difficult to diagnose and responses to specific treatments may vary between individuals. Therefore, there is an indisputable need for biomarkers for early diagnosis, prediction of disease exacerbations, monitoring the progression of disease, and for measuring responses to therapy. Genomic and proteomic studies have sought to understand the molecular basis of MS and find biomarker candidates. Advances in next-generation sequencing and mass-spectrometry techniques have yielded an unprecedented amount of genomic and proteomic data; yet, translation of the results into the clinic has been underwhelming. This has prompted the development of novel data science techniques for exploring these large datasets to identify biologically relevant relationships and ultimately point towards useful biomarkers. Herein we discuss optimization of omics study designs, advances in the generation of omics data, and systems biology approaches aimed at improving biomarker discovery and translation to the clinic for MS.
Collapse
Affiliation(s)
- Carol Chase Huizar
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh, UPMC Children's Hospital, Pittsburgh, PA, USA.
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
6
|
't Hart BA. Experimental autoimmune encephalomyelitis in the common marmoset: a translationally relevant model for the cause and course of multiple sclerosis. Primate Biol 2019; 6:17-58. [PMID: 32110715 PMCID: PMC7041540 DOI: 10.5194/pb-6-17-2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
Aging Western societies are facing an increasing prevalence of chronic
autoimmune-mediated inflammatory disorders (AIMIDs) for which treatments that are safe and effective are scarce. One of the
main reasons for this situation is the lack of animal models, which accurately replicate
clinical and pathological aspects of the human diseases. One important AIMID is the
neuroinflammatory disease multiple sclerosis (MS), for which the mouse experimental
autoimmune encephalomyelitis (EAE) model has been frequently used in preclinical
research. Despite some successes, there is a long list of experimental treatments that
have failed to reproduce promising effects observed in murine EAE models when they were
tested in the clinic. This frustrating situation indicates a wide validity gap between
mouse EAE and MS. This monography describes the development of an EAE model in nonhuman
primates, which may help to bridge the gap.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, the Netherlands.,Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, the Netherlands
| |
Collapse
|
7
|
't Hart BA, Laman JD, Kap YS. Merits and complexities of modeling multiple sclerosis in non-human primates: implications for drug discovery. Expert Opin Drug Discov 2018; 13:387-397. [PMID: 29465302 DOI: 10.1080/17460441.2018.1443075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The translation of scientific discoveries made in animal models into effective treatments for patients often fails, indicating that currently used disease models in preclinical research are insufficiently predictive for clinical success. An often-used model in the preclinical research of autoimmune neurological diseases, multiple sclerosis in particular, is experimental autoimmune encephalomyelitis (EAE). Most EAE models are based on genetically susceptible inbred/SPF mouse strains used at adolescent age (10-12 weeks), which lack exposure to genetic and microbial factors which shape the human immune system. Areas covered: Herein, the authors ask whether an EAE model in adult non-human primates from an outbred conventionally-housed colony could help bridge the translational gap between rodent EAE models and MS patients. Particularly, the authors discuss a novel and translationally relevant EAE model in common marmosets (Callithrix jacchus) that shares remarkable pathological similarity with MS. Expert opinion: The MS-like pathology in this model is caused by the interaction of effector memory T cells with B cells infected with the γ1-herpesvirus (CalHV3), both present in the pathogen-educated marmoset immune repertoire. The authors postulate that depletion of only the small subset (<0.05%) of CalHV3-infected B cells may be sufficient to limit chronic inflammatory demyelination.
Collapse
Affiliation(s)
- Bert A 't Hart
- a Department of Immunobiology , Biomedical Primate Research Centre , Rijswijk , The Netherlands.,b Department of Neuroscience , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Jon D Laman
- b Department of Neuroscience , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Yolanda S Kap
- a Department of Immunobiology , Biomedical Primate Research Centre , Rijswijk , The Netherlands
| |
Collapse
|
8
|
Glutamine/glutamate (Glx) concentration in prefrontal cortex predicts reversal learning performance in the marmoset. Behav Brain Res 2018; 346:11-15. [PMID: 29378291 DOI: 10.1016/j.bbr.2018.01.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/08/2018] [Accepted: 01/21/2018] [Indexed: 01/07/2023]
Abstract
This study used Magnetic Resonance Spectroscopy (MRS) to identify potential neurometabolitic markers of cognitive performance in male (n = 7) and female (n = 8) middle-aged (∼5 years old) common marmosets (Callithrix jacchus). Anesthetized marmosets were scanned with a 4.7 T/40 cm horizontal magnet equipped with 450 mT/m magnetic field gradients and a 20 G/cm magnetic field gradient insert, within 3 months of completing the CANTAB serial Reversal Learning task. Neurometabolite concentrations of N-Acetyl Asparate, Myo-Inositol, Choline, Phosphocreatine + creatine, Glutamate and Glutamine were acquired from a 3 mm3 voxel positioned in the Prefrontal Cortex (PFC). Males acquired the reversals (but not simple discriminations) faster than the females. Higher PFC Glx (glutamate + glutamine) concentration was associated with faster acquisition of the reversals. Interestingly, the correlation between cognitive performance and Glx was significant in males, but not in females. These results suggest that MRS is a useful tool to identify biochemical markers of cognitive performance in the healthy nonhuman primate brain and that biological sex modulates the relationship between neurochemical composition and cognition.
Collapse
|
9
|
Primate autoimmune disease models; lost for translation? Clin Transl Immunology 2016; 5:e122. [PMID: 28435673 PMCID: PMC5384286 DOI: 10.1038/cti.2016.82] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 01/06/2023] Open
Abstract
Replacement, reduction and refinement (the 3R's) are the leading principles in translational research with animals. To be useful a model should also be clinically Relevant (the 4th R). Work in a non-human primate model of multiple sclerosis, the experimental autoimmune encephalomyelitis model, reveals an inherent conflict among these 4R principles. The impossibility to harmonize all 4R's forms a major challenge when the model is applied in preclinical drug development.
Collapse
|
10
|
Maggi P, Sati P, Massacesi L. Magnetic resonance imaging of experimental autoimmune encephalomyelitis in the common marmoset. J Neuroimmunol 2016; 304:86-92. [PMID: 27743612 DOI: 10.1016/j.jneuroim.2016.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/28/2016] [Indexed: 12/26/2022]
Abstract
Magnetic resonance imaging (MRI) is an invaluable tool for the diagnosis and monitoring of patients with multiple sclerosis (MS) as well as for the study of the disease pathophysiology. Because of its strong clinical, radiological and histopathological similarities with the human disease, experimental autoimmune encephalomyelitis (EAE) in the common marmoset has been studied more intensively over the past several years. Here, we review the current knowledge on MRI in the marmoset EAE, and we outline the physiopathological significance and translational values of these studies with respect to MS. Accumulating evidences suggest that the application of conventional, as well as non-conventional, MRI techniques in the marmoset EAE is a promising approach to elucidate the pathological processes underlying the development of inflammatory demyelinated lesions in the central nervous system, potentially improving the identification and development of new therapeutics.
Collapse
Affiliation(s)
- Pietro Maggi
- Department of Neurology, Hôpital Erasme-Université libre de Bruxelles, Belgium
| | - Pascal Sati
- Translational Neuroradiology Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Luca Massacesi
- Department of Neurosciences, Drug Research, and Child Health, University of Florence, Florence, Italy
| |
Collapse
|
11
|
't Hart BA, Dunham J, Jagessar SA, Kap YS. The common marmoset (<i>Callithrix jacchus</i>): a relevant preclinical model of human (auto)immune-mediated inflammatory disease of the brain. Primate Biol 2016. [DOI: 10.5194/pb-3-9-2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Abstract. The increasing prevalence of chronic autoimmune-mediated inflammatory disorders (AIMIDs) in aging human populations creates a high unmet need for safe and effective medications. However, thus far the translation of pathogenic concepts developed in animal models into effective treatments for the patient has been notoriously difficult. The main reason is that currently used mouse-based animal models for the pipeline selection of promising new treatments were insufficiently predictive for clinical success. Regarding the high immunological similarity between human and non-human primates (NHPs), AIMID models in NHPs can help to bridge the translational gap between rodent and man. Here we will review the preclinical relevance of the experimental autoimmune encephalomyelitis (EAE) model in common marmosets (Callithrix jacchus), a small-bodied neotropical primate. EAE is a generic AIMID model projected on the human autoimmune neuro-inflammatory disease multiple sclerosis (MS).
Collapse
|
12
|
Methods for Testing Immunological Factors. DRUG DISCOVERY AND EVALUATION: PHARMACOLOGICAL ASSAYS 2016. [PMCID: PMC7122208 DOI: 10.1007/978-3-319-05392-9_45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Hypersensitivity reactions can be elicited by various factors: either immunologically induced, i.e., allergic reactions to natural or synthetic compounds mediated by IgE, or non-immunologically induced, i.e., activation of mediator release from cells through direct contact, without the induction of, or the mediation through immune responses. Mediators responsible for hypersensitivity reactions are released from mast cells. An important preformed mediator of allergic reactions found in these cells is histamine. Specific allergens or the calcium ionophore 48/80 induce release of histamine from mast cells. The histamine concentration can be determined with the o-phthalaldehyde reaction.
Collapse
|
13
|
Dang PT, Bui Q, D'Souza CS, Orian JM. Modelling MS: Chronic-Relapsing EAE in the NOD/Lt Mouse Strain. Curr Top Behav Neurosci 2015; 26:143-177. [PMID: 26126592 DOI: 10.1007/7854_2015_378] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Modelling complex disorders presents considerable challenges, and multiple sclerosis (MS) is no exception to this rule. The aetiology of MS is unknown, and its pathophysiology is poorly understood. Moreover, the last two decades have witnessed a dramatic revision of the long-held view of MS as an inflammatory demyelinating white matter disease. Instead, it is now regarded as a global central nervous system (CNS) disorder with a neurodegenerative component. Currently, there is no animal model recapitulating MS immunopathogenesis. Available models are based on autoimmune-mediated demyelination, denoted experimental autoimmune encephalomyelitis (EAE) or virally or chemically induced demyelination. Of these, the EAE model has been the most commonly used. It has been extensively improved since its first description and now exists as a number of variants, including genetically modified and humanized versions. Nonetheless, EAE is a distinct disease, and each variant models only certain facets of MS. Whilst the search for more refined MS models must continue, it is important to further explore where mechanisms underlying EAE provide proof-of-principle for those driving MS pathogenesis. EAE variants generated with the myelin component myelin oligodendrocyte glycoprotein (MOG) have emerged as the preferred ones, because in this particular variant disease is associated with both T- and B-cell effector mechanisms, together with demyelination. MOG-induced EAE in the non-obese diabetic (NOD) mouse strain exhibits a chronic-relapsing EAE clinical profile and high disease incidence. We describe the generation of this variant, its contribution to the understanding of MS immune and pathogenetic mechanisms and potential for evaluation of candidate therapies.
Collapse
Affiliation(s)
- Phuc T Dang
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Quyen Bui
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Claretta S D'Souza
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Jacqueline M Orian
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
14
|
Matthews PM, Comley R. Advances in the molecular imaging of multiple sclerosis. Expert Rev Clin Immunol 2014; 5:765-77. [DOI: 10.1586/eci.09.66] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
15
|
van der Greef J, van Wietmarschen H, van Ommen B, Verheij E. Looking back into the future: 30 years of metabolomics at TNO. MASS SPECTROMETRY REVIEWS 2013; 32:399-415. [PMID: 23630115 DOI: 10.1002/mas.21370] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 11/21/2012] [Accepted: 11/21/2012] [Indexed: 06/02/2023]
Abstract
Metabolites have played an essential role in our understanding of life, health, and disease for thousands of years. This domain became much more important after the concept of metabolism was discovered. In the 1950s, mass spectrometry was coupled to chromatography and made the technique more application-oriented and allowed the development of new profiling technologies. Since 1980, TNO has performed system-based metabolic profiling of body fluids, and combined with pattern recognition has led to many discoveries and contributed to the field known as metabolomics and systems biology. This review describes the development of related concepts and applications at TNO in the biomedical, pharmaceutical, nutritional, and microbiological fields, and provides an outlook for the future.
Collapse
|
16
|
Lutz NW, Fernandez C, Pellissier JF, Cozzone PJ, Béraud E. Cerebral biochemical pathways in experimental autoimmune encephalomyelitis and adjuvant arthritis: a comparative metabolomic study. PLoS One 2013; 8:e56101. [PMID: 23457507 PMCID: PMC3573043 DOI: 10.1371/journal.pone.0056101] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 01/09/2013] [Indexed: 01/17/2023] Open
Abstract
Many diseases, including brain disorders, are associated with perturbations of tissue metabolism. However, an often overlooked issue is the impact that inflammations outside the brain may have on brain metabolism. Our main goal was to study similarities and differences between brain metabolite profiles of animals suffering from experimental autoimmune encephalomyelitis (EAE) and adjuvant arthritis (AA) in Lewis rat models. Our principal objective was the determination of molecular protagonists involved in the metabolism underlying these diseases. EAE was induced by intraplantar injection of complete Freund’s adjuvant (CFA) and spinal-cord homogenate (SC-H), whereas AA was induced by CFA only. Naive rats served as controls (n = 9 for each group). Two weeks after inoculation, animals were sacrificed, and brains were removed and processed for metabolomic analysis by NMR spectroscopy or for immunohistochemistry. Interestingly, both inflammatory diseases caused similar, though not identical, changes in metabolites involved in regulation of brain cell size and membrane production: among the osmolytes, taurine and the neuronal marker, N-acetylaspartate, were decreased, and the astrocyte marker, myo-inositol, slightly increased in both inoculated groups compared with controls. Also ethanolamine-containing phospholipids, sources of inflammatory agents, and several glycolytic metabolites were increased in both inoculated groups. By contrast, the amino acids, aspartate and isoleucine, were less concentrated in CFA/SC-H and control vs. CFA rats. Our results suggest that inflammatory brain metabolite profiles may indicate the existence of either cerebral (EAE) or extra-cerebral (AA) inflammation. These inflammatory processes may act through distinct pathways that converge toward similar brain metabolic profiles. Our findings open new avenues for future studies aimed at demonstrating whether brain metabolic effects provoked by AA are pain/stress-mediated and/or due to the presence of systemic proinflammatory molecules. Regardless of the nature of these mechanisms, our findings may be of interest for future clinical studies, e.g. by in-vivo magnetic resonance spectroscopy.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Brain/metabolism
- Brain/pathology
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Freund's Adjuvant
- Metabolic Networks and Pathways
- Metabolomics
- Phospholipids/metabolism
- Rats
- Rats, Inbred Lew
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Water/metabolism
Collapse
Affiliation(s)
- Norbert W Lutz
- Center for Biological and Medical Magnetic Resonance CRMBM, National Center for Scientific Research Joint Research Unit 7339, Aix-Marseille University, Marseille, France.
| | | | | | | | | |
Collapse
|
17
|
't Hart BA, Abbott DH, Nakamura K, Fuchs E. The marmoset monkey: a multi-purpose preclinical and translational model of human biology and disease. Drug Discov Today 2012; 17:1160-5. [PMID: 22728226 DOI: 10.1016/j.drudis.2012.06.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 05/31/2012] [Accepted: 06/13/2012] [Indexed: 01/13/2023]
Abstract
The development of biologic molecules (monoclonal antibodies, cytokines, soluble receptors) as specific therapeutics for human disease creates a need for animal models in which safety and efficacy can be tested. Models in lower animal species are precluded when the reagents fail to recognize their targets, which is often the case in rats and mice. In this Feature article we will highlight the common marmoset, a small-bodied nonhuman primate (NHP), as a useful model in biomedical and preclinical translational research.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | | | | | | |
Collapse
|
18
|
't Hart BA, Jagessar SA, Kap YS, Brok HP. Preclinical models of multiple sclerosis in nonhuman primates. Expert Rev Clin Immunol 2010; 3:749-61. [PMID: 20477025 DOI: 10.1586/1744666x.3.5.749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Biotechnology has enabled the development of specifically acting therapies for immune-mediated inflammatory disorders (IMIDs) based on biological molecules. The high species specificity precludes safety and effectivity testing in lower species (mice and rats), thus creating a need for valid experimental models in nonhuman primates (NHPs). Here, we review the creation of relevant NHP model(s) for multiple sclerosis (MS), an IMID of the human CNS. We will also discuss how the model(s) can help in the translation of a scientific principle developed in lower species into a therapy for MS.
Collapse
Affiliation(s)
- Bert A 't Hart
- Biomedical Primate Research Centre and Erasmus Medical Centre Rotterdam, Rijswijk, The Netherlands.
| | | | | | | |
Collapse
|
19
|
't Hart BA, Hintzen RQ, Laman JD. Multiple sclerosis - a response-to-damage model. Trends Mol Med 2009; 15:235-44. [PMID: 19451035 DOI: 10.1016/j.molmed.2009.04.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Revised: 04/06/2009] [Accepted: 04/06/2009] [Indexed: 11/18/2022]
Abstract
According to a widely supported but unproven concept, the autoimmune mechanisms that drive neuroinflammation in multiple sclerosis (MS) are triggered by virus infection. However, a direct viral trigger of MS has not been identified. MS models in non-human primates suggest that lifelong asymptomatic infection with certain herpesviruses (e.g. cytomegalovirus) creates a repertoire of potentially autoreactive memory T cells. When these are exposed to antigens released after central nervous system injury as a consequence of an unknown pathogenic event, they are reactivated and induce autoimmune neurological disease. This response-to-damage of antiviral memory cells can take place years after the initiating infection. Consequently, elucidating the anti-herpesvirus T-cell repertoire might provide new targets for preventive diagnosis and therapy.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Center, Lange Kleiweg 139, 2288 GJ Rijswijk, The Netherlands.
| | | | | |
Collapse
|
20
|
't Hart BA, Hintzen RQ, Laman JD. Preclinical assessment of therapeutic antibodies against human CD40 and human interleukin-12/23p40 in a nonhuman primate model of multiple sclerosis. NEURODEGENER DIS 2008; 5:38-52. [PMID: 18075274 DOI: 10.1159/000109937] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Accepted: 01/11/2007] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Proinflammatory cytokines, such as interleukin (IL)-12 and IL-23, and costimulatory molecules on antigen-presenting cells (APC), such as CD40, are critical to autoreactive T cell activation by APC, and hence, are considered relevant targets of therapy for immune-mediated inflammatory diseases (IMID). OBJECTIVE The current review discusses the preclinical evaluation of two novel immunotherapeutic monoclonal antibodies (mAbs), one directed against human IL-12/23p40 and the other against CD40. As the antibodies only recognize their target molecule in primates, the efficacy could not be tested in rodent models. RESULTS As a preclinical IMID model for the in vivo evaluation of both mAbs, we have used the experimental autoimmune/allergic encephalomyelitis (EAE) model in common marmoset monkeys (Callithrix jacchus). Both mAbs show beneficial activities in the EAE model when administered early in disease development as well as after the onset of brain inflammation. The treatment effects were evaluated using a combination of quantitative magnetic resonance imaging and a series of ex vivo and immunopathological evaluations. CONCLUSION The promising effects during ongoing disease in a relevant preclinical IMID model illustrate the potential of these two antibodies as treatment of IMID, in particular for multiple sclerosis on which disease EAE has been modeled.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | | | | |
Collapse
|
21
|
Blezer ELA, Bauer J, Brok HPM, Nicolay K, 't Hart BA. Quantitative MRI-pathology correlations of brain white matter lesions developing in a non-human primate model of multiple sclerosis. NMR IN BIOMEDICINE 2007; 20:90-103. [PMID: 16948176 DOI: 10.1002/nbm.1085] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) induced with recombinant human myelin/oligodendrocyte glycoprotein in the common marmoset is a useful preclinical model of multiple sclerosis in which white matter lesions can be well visualized with MRI. In this study we characterized lesion progression with quantitative in vivo MRI (4.7 T; T(1) relaxation time +/- Gd-DTPA; T(2) relaxation time; magnetization transfer ratio, MTR, imaging) and correlated end stage MRI presentation with quantitative ex vivo MRI (formaldehyde fixed brains; T(1) and T(2) relaxation times; MTR) and histology. The histopathological characterization included axonal density measurements and the numeric quantification of infiltrated macrophages expressing markers for early active [luxol fast blue (LFB) or migration inhibition factor-related protein-14 positive] or late active/inactive [periodic acid Schiff (PAS) positive] demyelinating lesion. MRI experiments were done every two weeks until the monkeys were sacrificed with severe EAE-related motor deficits. Compared with the normal appearing white matter, lesions showed an initial increase in T(1) relaxation times, leakage of Gd-DTPA and decrease in MTR values. The progressive enlargement of lesions was associated with stabilized T(1) values, while T(2) initially increased and stabilized thereafter and MTR remained decreased. Gd-DTPA leakage was highly variable throughout the experiment. MRI characteristics of the cortex and (normal appearing) white matter did not change during the experiment. We observed that in vivo MTR values correlated positively with the number of early active (LFB+) and negatively with late active (PAS+) macrophages. Ex vivo MTR and relaxation times correlated positively with the number of PAS-positive macrophages. None of the investigated MRI parameters correlated with axonal density.
Collapse
Affiliation(s)
- Erwin L A Blezer
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|
22
|
Merkler D, Böscke R, Schmelting B, Czéh B, Fuchs E, Brück W, Stadelmann C. Differential macrophage/microglia activation in neocortical EAE lesions in the marmoset monkey. Brain Pathol 2006; 16:117-23. [PMID: 16768751 PMCID: PMC8095787 DOI: 10.1111/j.1750-3639.2006.00004.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Recent studies revealed an important involvement of the cerebral cortex in multiple sclerosis (MS) patients. Cortical lesions in MS were reported to be less inflammatory and to show less structural damage than white matter lesions. Animal models reflecting the histopathological hallmarks of cortical demyelinated lesions in MS are sparse. Induction of experimental autoimmune encephalomyelitis (EAE) in the common marmoset has turned out to be an attractive non-human-primate model for MS. In the present study we investigated the presence and detailed cellular composition of cortical inflammatory demyelinating pathology in the common marmoset upon immunization with myelin oligodendrocyte glycoprotein (MOG). Extensive cortical demyelination reflecting the topographically distinct cortical lesion types in MS patients was revealed by immunohistochemistry for myelin basic protein (MBP). We explored the density of T- and B-lymphocytes, MHC-II expressing macrophages/microglia cells and early activated macrophages (MRP14) at perivascular and parenchymal lesions sites in neocortex and subcortical white matter. Despite a similar density of perivascular inflammatory infiltrates in the demyelinated neocortex, a considerable lower fraction of macrophages was found to express MRP14 in the neocortex indicating a different activation pattern in cortical compared with white matter lesions. Furthermore, cortical EAE lesions in marmoset monkeys revealed immunoglobulin leakage and complement component C9 deposition in intracortical but not subpial demyelination. Our findings indicate that the inflammatory response, especially macrophage and microglia activation, may be regulated differently in gray matter areas in primate brain.
Collapse
Affiliation(s)
- Doron Merkler
- Department of Neuropathology, Georg-August University Göttingen, Göttingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
23
|
Petibois C, Déléris G. Chemical mapping of tumor progression by FT-IR imaging: towards molecular histopathology. Trends Biotechnol 2006; 24:455-62. [PMID: 16935373 DOI: 10.1016/j.tibtech.2006.08.005] [Citation(s) in RCA: 213] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2006] [Revised: 07/12/2006] [Accepted: 08/15/2006] [Indexed: 11/28/2022]
Abstract
Fourier-transform infrared (FT-IR) spectro-imaging enables global analysis of samples, with resolution close to the cellular level. Recent studies have shown that FT-IR imaging enables determination of the biodistribution of several molecules of interest (carbohydrates, lipids, proteins) for tissue analysis without pre-analytical modification of the sample such as staining. Molecular structure information is also available from the same analysis, notably for protein secondary structure and fatty acyl chain peroxidation level. Thus, several cancer markers can be identified from FT-IR tissue images, enabling accurate discrimination between healthy and tumor areas. FT-IR imaging applications are now able to provide unique chemical and morphological information about tissue status. With the fast image acquisition provided by modern mid-infrared imaging systems, it is now envisaged to analyze cerebral tumor exereses in delays compatible with neurosurgery. Accordingly, we propose to take FT-IR imaging into consideration for the development of new molecular histopathology tools.
Collapse
Affiliation(s)
- Cyril Petibois
- Université Victor Segalen Bordeaux 2, CNRS UMR 5084, CNAB, Bio-Organic Chemistry Group, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France.
| | | |
Collapse
|
24
|
Boretius S, Schmelting B, Watanabe T, Merkler D, Tammer R, Czéh B, Michaelis T, Frahm J, Fuchs E. Monitoring of EAE onset and progression in the common marmoset monkey by sequential high-resolution 3D MRI. NMR IN BIOMEDICINE 2006; 19:41-9. [PMID: 16408325 DOI: 10.1002/nbm.999] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) induced by myelin-oligodendrocyte glycoprotein (MOG) in common marmosets (Callithrix jacchus) is a model for multiple sclerosis. Here, EAE was induced in four common marmosets by 250-300 microg recombinant rat MOG. In addition to a detailed disability scoring, T2- and T1-weighted high-resolution 3D MRI was performed to assess the onset and development of cerebral lesions. The findings were confirmed by histopathology in all animals. Although the animals exhibited a large heterogeneity with regard to onset and localization of lesions and also to disease duration and severity of disability signs, none of the animals revealed any evidence of recovery. A specification of the disability scoring system to account for different aspects of the disease led to a good concurrence of the first MRI-detectable lesion and the onset of central nervous system (CNS) symptoms. The results suggest that MRI monitoring of white matter lesions in conjunction with disability scores that focus on CNS symptoms may be a suitable method to evaluate novel therapeutic interventions even in the presence of pronounced interindividual heterogeneity.
Collapse
Affiliation(s)
- Susann Boretius
- Biomedizinische NMR Forschungs GmbH am Max-Planck-Institut für Biophysikalische Chemie, 37070 Göttingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
't Hart BA, Smith P, Amor S, Strijkers GJ, Blezer ELA. MRI-guided immunotherapy development for multiple sclerosis in a primate. Drug Discov Today 2006; 11:58-66. [PMID: 16478692 DOI: 10.1016/s1359-6446(05)03673-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Multiple sclerosis is a serious neurological disease that affects 1 in 1000 young adults in Europe and the USA. The development of an effective therapy for this enigmatic disease is plagued by the failure of many treatments to reproduce in patients the promising effects observed in animal models. This review describes a new preclinical model in a non-human primate that might help to bridge the gap between currently used animal models and the patients.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | | | | | | | | |
Collapse
|
26
|
Abstract
Multiple sclerosis (MS) is both a complex and chronic neurological disease of the CNS. This poses unique challenges for drug discovery in terms of delineating specific targets related to disease mechanisms and developing safe and effective molecules for clinical application. Preclinical animal models of MS provide the necessary test bed for evaluating the effects of novel therapeutic strategies. Because the clinical manifestations and pathological consequences of disease vary dramatically from individual to individual, as well as treatment response to existing therapies, this creates a significant research endeavor in terms of translating preclinical methodologies to the clinical domain. Potentially exciting treatments have emerged in the form of natalizumab (Tysabri), an alpha4 integrin antagonist, and more recently FTY720, a sphinogosine-1 phosphate receptor modulator, providing a compelling proof-of-principle from bench to bedside. However, further research is required to discharge safety concerns associated with these therapeutic avenues. Future prospects in the guise of disease-modifying therapies that target the inflammatory and neurodegenerative components of disease have come to the forefront of preclinical research with the sole aim of reducing the underlying irreversible progressive disability of MS. Significant progress with novel therapies will be made by implementing biomarker strategies that extrapolate robustly from animal models to the divergent patient populations of MS. The future therapeutic options for MS will depend on improvements in understanding the precise factors involved in disease onset and progression and subsequently the development of oral therapeutics that translate sustained benefit from the preclinical context into clinical reality.
Collapse
Affiliation(s)
- David J Virley
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Pharmaceuticals, Harlow, Essex CM19 5AW, United Kingdom.
| |
Collapse
|
27
|
't Hart BA, Brok HPM, Remarque E, Benson J, Treacy G, Amor S, Hintzen RQ, Laman JD, Bauer J, Blezer ELA. Suppression of Ongoing Disease in a Nonhuman Primate Model of Multiple Sclerosis by a Human-Anti-Human IL-12p40 Antibody. THE JOURNAL OF IMMUNOLOGY 2005; 175:4761-8. [PMID: 16177124 DOI: 10.4049/jimmunol.175.7.4761] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
IL-12p40 is a shared subunit of two cytokines with overlapping activities in the induction of autoreactive Th1 cells and therefore a potential target of therapy in Th1-mediated diseases. We have examined whether ongoing disease in a nonhuman primate model of multiple sclerosis (MS) can be suppressed with a new human IgG1kappa Ab against human IL-12p40. Lesions developing in the brain white matter were visualized and characterized with standard magnetic resonance imaging techniques. To reflect the treatment of MS patients, treatment with the Ab was initiated after active brain white matter lesions were detected in T2-weighted images. In placebo-treated control monkeys we observed the expected progressive increase in the total T2 lesion volume and markedly increased T2 relaxation times, a magnetic resonance imaging marker of inflammation. In contrast, in monkeys treated with anti-IL-12p40 Ab, changes in the total T2 lesion volume and T2 relaxation times were significantly suppressed. Moreover, the time interval to serious neurological deficit was delayed from 31 +/- 10 to 64 +/- 20 days (odds ratio, 0.312). These results, in a disease model with high similarity to MS, are important for ongoing and planned trials of therapies that target IL-12 and/or IL-23.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Brain/pathology
- Callithrix
- Disease Models, Animal
- Disease Progression
- Drug Evaluation, Preclinical
- Encephalomyelitis, Autoimmune, Experimental/diagnosis
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Humans
- Interleukin-12/immunology
- Interleukin-12 Subunit p40
- Magnetic Resonance Imaging
- Male
- Multiple Sclerosis/diagnosis
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Multiple Sclerosis/prevention & control
- Protein Subunits/immunology
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW This review will focus on recent developments in multiple sclerosis (MS) pathology with particular emphasis on the patterns and mediators of lesion formation in MS, the mechanisms of oligodendrocyte and axon damage and the magnetic resonance imaging-pathological correlation of MS lesions. RECENT STUDIES The inflammatory cascade in the MS plaque has been characterized in more detail, and other factors such as hypoxia-like injury or excitotoxicity, besides immunological effector mechanisms, have been found to play a role in MS pathogenesis. Cortical demyelination and mechanisms of neuroaxonal damage are discussed in detail. The radiological correlate of basic histopathologic findings is being approached with quantitative methods. Similar quantitative approaches are used in MS-gene expression studies that compare patterns of gene expression in different lesion areas. SUMMARY These studies will lead to better understanding of the pathogenesis of MS lesions and will hopefully identify new therapeutic targets to modulate inflammation, support remyelination and protect axons and neurons in MS.
Collapse
Affiliation(s)
- Wolfgang Brück
- Department of Neuropathology, Georg-August University Göttingen, Göttingen, Germany.
| | | |
Collapse
|
29
|
't Hart BA, Bauer J, Brok HPM, Amor S. Non-human primate models of experimental autoimmune encephalomyelitis: Variations on a theme. J Neuroimmunol 2005; 168:1-12. [PMID: 16023737 DOI: 10.1016/j.jneuroim.2005.05.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Revised: 05/26/2005] [Accepted: 05/27/2005] [Indexed: 12/31/2022]
Abstract
Despite years of intensive research into multiple sclerosis (MS) scientists have not yet succeeded in developing an absolute therapy for the treatment of this disabling disease of the human central nervous system. The wide immunological gap between inbred rodent strains and the heterogeneous human population is probably the single most important factor that hampers the translation of scientific principles developed in rodents into effective therapies for MS. Because of the closer immunological proximity to humans, non-human primates provide useful experimental models that may help to bridge this gap. Here we review the models of experimental autoimmune encephalomyelitis in rhesus macaques and common marmosets. We will discuss the salient points of the models and suggest how these may represent the spectrum of inflammatory demyelinating diseases of the central nervous system in humans.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | | | | | | |
Collapse
|
30
|
't Hart BA, Blezer ELA, Brok HPM, Boon L, de Boer M, Bauer J, Laman JD. Treatment with chimeric anti-human CD40 antibody suppresses MRI-detectable inflammation and enlargement of pre-existing brain lesions in common marmosets affected by MOG-induced EAE. J Neuroimmunol 2005; 163:31-9. [PMID: 15885306 DOI: 10.1016/j.jneuroim.2005.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2004] [Revised: 02/07/2005] [Accepted: 02/07/2005] [Indexed: 11/17/2022]
Abstract
Common marmosets, a Neotropical monkey species, are protected against clinical and neuropathological consequences of experimentally induced autoimmune encephalomyelitis (EAE) by prophylactic treatment with ch5D12, a humanized antagonist antibody against human CD40. In the current study we have tested whether ch5D12 acts therapeutically against the enlargement and inflammatory activity of existing (brain) white matter lesions using serial magnetic resonance imaging (MRI). The results show in all PBS treated monkeys (n=4) a rapid enlargement of T2 lesions together with an increment of the T2 signal intensity due to inflammatory edema. Treatment with ch5D12 delayed the enlargement of T2 lesions in 2 out of 3 tested monkeys while in 3 out of 3 monkeys the T2 signal increment of lesions was suppressed. In conjunction with previously published data on the clinical benefit of anti-CD40 treatment in the marmoset EAE model, the current findings support antibody-mediated blockade of CD40 interaction with its ligand CD154 as a potential treatment of MS.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
31
|
Rudin M, Beckmann N, Rausch M. Evaluation of drug candidates: efficacy readouts during lead optimization. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2005; 62:185-255. [PMID: 16329258 DOI: 10.1007/3-7643-7426-8_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Affiliation(s)
- Markus Rudin
- Institute for Biomedical Engineering, University of Zurich/ETH Zurich, Switzerland.
| | | | | |
Collapse
|
32
|
't Hart BA, Laman JD, Bauer J, Blezer E, van Kooyk Y, Hintzen RQ. Modelling of multiple sclerosis: lessons learned in a non-human primate. Lancet Neurol 2004; 3:588-97. [PMID: 15380155 DOI: 10.1016/s1474-4422(04)00879-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The many, highly specific, biological therapies for immune-based diseases create a need for valid preclinical animal models. The wide immunological gap between human beings and laboratory mouse or rat models makes many disease models in these species invalid. In this review, we report a non-human-primate model of chronic multiple sclerosis (MS)-experimental autoimmune encephalitis (EAE) in the common marmoset (Callithrix jacchus)-that can help bridge this wide gap. The genetic and immunological similarity of marmosets and human beings and the clinical and neuropathological similarity of the EAE model to MS provide a unique experimental platform for research into basic immunopathogenetic mechanisms and for the development of more effective treatments for MS.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, Netherlands.
| | | | | | | | | | | |
Collapse
|
33
|
't Hart BA, Amor S, Jonker M. Evaluating the validity of animal models for research into therapies for immune-based disorders. Drug Discov Today 2004; 9:517-24. [PMID: 15183159 DOI: 10.1016/s1359-6446(04)03112-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The last few decades of the 20th century have shown an intensified search for safer and more effective medications against chronic diseases that burden ageing societies of the western world. The impressive development of biotechnological production techniques has greatly facilitated the pharmaceutical development of relatively non-toxic biological molecules. However, despite the huge investments, only a few effective therapies for immune-based diseases have reached the clinic. In this article we use examples from monoclonal antibody trials to discuss the validity and predictive strength of the animal models currently used for the development of effective therapies.
Collapse
MESH Headings
- Adjuvants, Immunologic/therapeutic use
- Animals
- Antibodies, Monoclonal/therapeutic use
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Disease Models, Animal
- Drug Evaluation, Preclinical/methods
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Immune System Diseases/immunology
- Immune System Diseases/therapy
- Immunotherapy/methods
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Organ Transplantation
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | | | | |
Collapse
|