1
|
Lazarević M, Stanisavljević S, Nikolovski N, Dimitrijević M, Miljković Đ. Complete Freund's adjuvant as a confounding factor in multiple sclerosis research. Front Immunol 2024; 15:1353865. [PMID: 38426111 PMCID: PMC10902151 DOI: 10.3389/fimmu.2024.1353865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Complete Freund's adjuvant (CFA) is used as a standard adjuvant for the induction of experimental autoimmune encephalomyelitis (EAE), the most commonly used animal model in multiple sclerosis studies. Still, CFA induces glial activation and neuroinflammation on its own and provokes pain. In addition, as CFA contains Mycobacteria, an immune response against bacterial antigens is induced in parallel to the response against central nervous system antigens. Thus, CFA can be considered as a confounding factor in multiple sclerosis-related studies performed on EAE. Here, we discuss the effects of CFA in EAE in detail and present EAE variants induced in experimental animals without the use of CFA. We put forward CFA-free EAE variants as valuable tools for studying multiple sclerosis pathogenesis and therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
2
|
Doelman W, Reijnen RC, Dijksman N, Janssen APA, van Driel N, 't Hart BA, Philippens I, Araman C, Baron W, van Kasteren SI. Citrullinated human and murine MOG 35-55 display distinct biophysical and biochemical behavior. J Biol Chem 2023; 299:103065. [PMID: 36841486 PMCID: PMC10060747 DOI: 10.1016/j.jbc.2023.103065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023] Open
Abstract
The peptide spanning residues 35 to 55 of the protein myelin oligodendrocyte glycoprotein (MOG) has been studied extensively in its role as a key autoantigen in the neuroinflammatory autoimmune disease multiple sclerosis. Rodents and nonhuman primate species immunized with this peptide develop a neuroinflammatory condition called experimental autoimmune encephalomyelitis, often used as a model for multiple sclerosis. Over the last decade, the role of citrullination of this antigen in the disease onset and progression has come under increased scrutiny. We recently reported on the ability of these citrullinated MOG35-55 peptides to aggregate in an amyloid-like fashion, suggesting a new potential pathogenic mechanism underlying this disease. The immunodominant region of MOG is highly conserved between species, with the only difference between the murine and human protein, a polymorphism on position 42, which is serine in mice and proline for humans. Here, we show that the biophysical and biochemical behavior we previously observed for citrullinated murine MOG35-55 is fundamentally different for human and mouse MOG35-55. The citrullinated human peptides do not show amyloid-like behavior under the conditions where the murine peptides do. Moreover, we tested the ability of these peptides to stimulate lymphocytes derived from MOG immunized marmoset monkeys. While the citrullinated murine peptides did not produce a proliferative response, one of the citrullinated human peptides did. We postulate that this unexpected difference is caused by disparate antigen processing. Taken together, our results suggest that further study on the role of citrullination in MOG-induced experimental autoimmune encephalomyelitis is necessary.
Collapse
Affiliation(s)
- W Doelman
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - R C Reijnen
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - N Dijksman
- Section Molecular Neurobiology, Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - A P A Janssen
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - N van Driel
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, the Netherlands
| | - B A 't Hart
- Section Molecular Neurobiology, Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - I Philippens
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, the Netherlands
| | - C Araman
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - W Baron
- Section Molecular Neurobiology, Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - S I van Kasteren
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands.
| |
Collapse
|
3
|
Laman JD. Cutting edge technologies in chronic inflammation research. Exp Dermatol 2022; 31 Suppl 1:17-21. [PMID: 36059185 PMCID: PMC9539701 DOI: 10.1111/exd.14648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/27/2022]
Abstract
This concise review provides the broad background and selection from the literature for a Keynote lecture at EHSF 2022 on state of the art technologies in inflammation research, with an emphasis on disease of the skin and the nervous system. The value of ex vivo skin explant models is discussed, as well as the innovative use of animal models, wherein the crucial roles of antigen experience and "wild" microbiota are emphasized. Spectral flow cytometry allowing large surface marker panels to be explored is touched upon, as well as multiplex technology for cytokines and other analytes important for inflammation and tissue damage. Single-cell sequencing and in situ transcriptomics (spatial profiling) now provide exciting granular information on functional cell subsets, interactions and plasticity. A selection of novel research and diagnostic tools for antibodies against linear peptides or gangliosides is presented. Finally, the review discusses a new anti-inflammatory strategy against skin inflammation with a panel of protease inhibitors derived from the protein fraction of industrial starch potatoes.
Collapse
Affiliation(s)
- Jon D Laman
- Department of Pathology and Medical Biology, University Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| |
Collapse
|
4
|
Multiple sclerosis and drug discovery: A work of translation. EBioMedicine 2021; 68:103392. [PMID: 34044219 PMCID: PMC8245896 DOI: 10.1016/j.ebiom.2021.103392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/26/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023] Open
Abstract
Multiple sclerosis (MS) is after trauma the most important neurological disease in young adults, affecting 1 per 1000 individuals. With currently available medications, most of these targeting the immune system, satisfactory results have been obtained in patients with relapsing MS, but these can have serious adverse effects. Moreover, despite some promising developments, such as with B cell targeting therapies or sphingosine-1-phosphate modulating drugs, there still is a high unmet need of safe drugs with broad efficacy in patients with progressive MS. Despite substantial investments and intensive preclinical research, the proportion of promising lead compounds that reaches the approved drug status remains disappointingly low. One cause lies in the poor predictive validity of MS animal models used in the translation of pathogenic mechanisms into safe and effective treatments for the patient. This disturbing situation has raised criticism against the relevance of animal models used in preclinical research and calls for improvement of these models. This publication presents a potentially useful strategy to enhance the predictive validity of MS animal models, namely, to analyze the causes of failure in forward translation (lab to clinic) via reverse translation (clinic to lab). Through this strategy new insights can be gained that can help generate more valid MS models.
Collapse
|
5
|
Complete Freund's adjuvant-free experimental autoimmune encephalomyelitis in Dark Agouti rats is a valuable tool for multiple sclerosis studies. J Neuroimmunol 2021; 354:577547. [PMID: 33765502 DOI: 10.1016/j.jneuroim.2021.577547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/02/2021] [Accepted: 03/12/2021] [Indexed: 12/21/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is classically induced with complete Freund's adjuvant (CFA). The immune response against CFA has a confounding influence on the translational capacity of EAE as a multiple sclerosis model. Here, we compare clinical, cellular and molecular properties between syngeneic spinal cord homogenate (SCH)- and SCH + CFA-immunized Dark Agouti rats. EAE signs were observed earlier and the cumulative clinical score was higher without CFA. Also, a higher number of immune cells infiltrates in the spinal cords was noticed at the peak of EAE without CFA. High spinal cord abundance of CD8+CD11bc+MHC class II+ cells was detected in SCH-immunized rats. Myelin basic protein -specific response can be elicited in the cells from the lymph nodes draining the site of SCH immunization. This CFA-free EAE is a reliable multiple sclerosis model.
Collapse
|
6
|
Doelman W, Marqvorsen MHS, Chiodo F, Bruijns SCM, van der Marel GA, van Kooyk Y, van Kasteren SI, Araman C. Synthesis of Asparagine Derivatives Harboring a Lewis X Type DC-SIGN Ligand and Evaluation of their Impact on Immunomodulation in Multiple Sclerosis. Chemistry 2020; 27:2742-2752. [PMID: 33090600 PMCID: PMC7898482 DOI: 10.1002/chem.202004076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Indexed: 01/13/2023]
Abstract
The protein myelin oligodendrocyte glycoprotein (MOG) is a key component of myelin and an autoantigen in the disease multiple sclerosis (MS). Post‐translational N‐glycosylation of Asn31 of MOG seems to play a key role in modulating the immune response towards myelin. This is mediated by the interaction of Lewis‐type glycan structures in the N‐glycan of MOG with the DC‐SIGN receptor on dendritic cells (DCs). Here, we report the synthesis of an unnatural Lewis X (LeX)‐containing Fmoc‐SPPS‐compatible asparagine building block (SPPS=solid‐phase peptide synthesis), as well as asparagine building blocks containing two LeX‐derived oligosaccharides: LacNAc and Fucα1‐3GlcNAc. These building blocks were used for the glycosylation of the immunodominant portion of MOG (MOG31‐55) and analyzed with respect to their ability to bind to DC‐SIGN in different biological setups, as well as their ability to inhibit the citrullination‐induced aggregation of MOG31‐55. Finally, a cytokine secretion assay was carried out on human monocyte‐derived DCs, which showed the ability of the neoglycopeptide decorated with a single LeX to alter the balance of pro‐ and anti‐inflammatory cytokines, inducing a tolerogenic response.
Collapse
Affiliation(s)
- Ward Doelman
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Mikkel H S Marqvorsen
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Fabrizio Chiodo
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC-Location Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Sven C M Bruijns
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC-Location Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Gijsbert A van der Marel
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC-Location Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Sander I van Kasteren
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Can Araman
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| |
Collapse
|
7
|
Armangue T, Capobianco M, de Chalus A, Laetitia G, Deiva K. E.U. paediatric MOG consortium consensus: Part 3 - Biomarkers of paediatric myelin oligodendrocyte glycoprotein antibody-associated disorders. Eur J Paediatr Neurol 2020; 29:22-31. [PMID: 33191096 DOI: 10.1016/j.ejpn.2020.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022]
Abstract
A first episode of acquired demyelinating disorder (ADS) in children is a diagnostic challenge as different diseases can express similar clinical features. Recently, antibodies against myelin oligodendrocyte glycoprotein (MOG) have emerged as a new ADS biomarker, which clearly allow the identification of monophasic and relapsing ADS forms different from MS predominantly in children. Due to the novelty of this antibody there are still challenges and controversies about its pathogenicity and best technique to detect it. In this manuscript we will discuss the recommendations and caveats on MOG antibody assays, role in the pathogenesis, and additionally discuss the usefulness of other potential new biomarkers in MOG-antibody associated disorders (MOGAD).
Collapse
Affiliation(s)
- Thaís Armangue
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Pediatric Neuroimmunology Unit, Neurology Department, Sant Joan de Déu (SJD) Children's Hospital, University of Barcelona, Barcelona, Spain.
| | - Marco Capobianco
- Department of Neurology and Regional Multiple Sclerosis Centre, University Hospital San Luigi Gonzaga, Orbassano, Italy
| | - Aliénor de Chalus
- Assistance Publique-Hôpitaux de Paris, Pediatric Neurology Department, University Hospitals Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Giorgi Laetitia
- Assistance Publique-Hôpitaux de Paris, Pediatric Neurology Department, University Hospitals Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Kumaran Deiva
- Assistance Publique-Hôpitaux de Paris, Pediatric Neurology Department, University Hospitals Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France; French Reference Network of Rare Inflammatory Brain and Spinal Diseases, Le Kremlin Bicêtre, France and European Reference Network-RITA, France
| | | |
Collapse
|
8
|
't Hart BA. A Tolerogenic Role of Cathepsin G in a Primate Model of Multiple Sclerosis: Abrogation by Epstein-Barr Virus Infection. Arch Immunol Ther Exp (Warsz) 2020; 68:21. [PMID: 32556812 PMCID: PMC7299916 DOI: 10.1007/s00005-020-00587-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 06/11/2020] [Indexed: 11/25/2022]
Abstract
Using a non-human primate model of the autoimmune neuroinflammatory disease multiple sclerosis (MS), we have unraveled the role of B cells in the making and breaking of immune tolerance against central nervous system myelin. It is discussed here that B cells prevent the activation of strongly pathogenic T cells present in the naïve repertoire, which are directed against the immunodominant myelin antigen MOG (myelin oligodendrocyte glycoprotein). Prevention occurs via destructive processing of a critical epitope (MOG34-56) through the lysosomal serine protease cathepsin G. This effective tolerance mechanism is abrogated when the B cells are infected with Epstein–Barr virus, a ubiquitous γ1-herpesvirus that entails the strongest non-genetic risk factor for MS.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center, Groningen, The Netherlands. .,Department of Anatomy and Neurosciences, VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
'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
|
10
|
Leibovitch EC, Caruso B, Ha SK, Schindler MK, Lee NJ, Luciano NJ, Billioux BJ, Guy JR, Yen C, Sati P, Silva AC, Reich DS, Jacobson S. Herpesvirus trigger accelerates neuroinflammation in a nonhuman primate model of multiple sclerosis. Proc Natl Acad Sci U S A 2018; 115:11292-11297. [PMID: 30322946 PMCID: PMC6217390 DOI: 10.1073/pnas.1811974115] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Pathogens, particularly human herpesviruses (HHVs), are implicated as triggers of disease onset/progression in multiple sclerosis (MS) and other neuroinflammatory disorders. However, the time between viral acquisition in childhood and disease onset in adulthood complicates the study of this association. Using nonhuman primates, we demonstrate that intranasal inoculations with HHV-6A and HHV-6B accelerate an MS-like neuroinflammatory disease, experimental autoimmune encephalomyelitis (EAE). Although animals inoculated intranasally with HHV-6 (virus/EAE marmosets) were asymptomatic, they exhibited significantly accelerated clinical EAE compared with control animals. Expansion of a proinflammatory CD8 subset correlated with post-EAE survival in virus/EAE marmosets, suggesting that a peripheral (viral?) antigen-driven expansion may have occurred post-EAE induction. HHV-6 viral antigen in virus/EAE marmosets was markedly elevated and concentrated in brain lesions, similar to previously reported localizations of HHV-6 in MS brain lesions. Collectively, we demonstrate that asymptomatic intranasal viral acquisition accelerates subsequent neuroinflammation in a nonhuman primate model of MS.
Collapse
Affiliation(s)
- Emily C Leibovitch
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Breanna Caruso
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Seung Kwon Ha
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Matthew K Schindler
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Nathanael J Lee
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Nicholas J Luciano
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Bridgette J Billioux
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Joseph R Guy
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Cecil Yen
- Cerebral Microcirculation Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Pascal Sati
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Afonso C Silva
- Cerebral Microcirculation Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Steven Jacobson
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
11
|
Kap YS, Bus-Spoor C, van Driel N, Dubbelaar ML, Grit C, Kooistra SM, Fagrouch ZC, Verschoor EJ, Bauer J, Eggen BJL, Harmsen HJM, Laman JD, 't Hart BA. Targeted Diet Modification Reduces Multiple Sclerosis-like Disease in Adult Marmoset Monkeys from an Outbred Colony. THE JOURNAL OF IMMUNOLOGY 2018; 201:3229-3243. [PMID: 30341184 DOI: 10.4049/jimmunol.1800822] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/25/2018] [Indexed: 01/20/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) in common marmosets is a translationally relevant model of the chronic neurologic disease multiple sclerosis. Following the introduction of a new dietary supplement in our purpose-bred marmoset colony, the percentage of marmosets in which clinically evident EAE could be induced by sensitization against recombinant human myelin oligodendrocyte glycoprotein in IFA decreased from 100 to 65%. The reduced EAE susceptibility after the dietary change coincided with reduced Callitrichine herpesvirus 3 expression in the colony, an EBV-related γ1-herpesvirus associated with EAE. We then investigated, in a controlled study in marmoset twins, which disease-relevant parameters were affected by the dietary change. The selected twins had been raised on the new diet for at least 12 mo prior to the study. In twin siblings reverted to the original diet 8 wk prior to EAE induction, 100% disease prevalence (eight out of eight) was restored, whereas in siblings remaining on the new diet the EAE prevalence was 75% (six out of eight). Spinal cord demyelination, a classical hallmark of the disease, was significantly lower in new-diet monkeys than in monkeys reverted to the original diet. In new-diet monkeys, the proinflammatory T cell response to recombinant human myelin oligodendrocyte glycoprotein was significantly reduced, and RNA-sequencing revealed reduced apoptosis and enhanced myelination in the brain. Systematic typing of the marmoset gut microbiota using 16S rRNA sequencing demonstrated a unique, Bifidobacteria-dominated composition, which changed after disease induction. In conclusion, targeted dietary intervention exerts positive effects on EAE-related parameters in multiple compartments of the marmoset's gut-immune-CNS axis.
Collapse
Affiliation(s)
- Yolanda S Kap
- Department of Immunobiology, Biomedical Primate Research Centre, 2280 GH Rijswijk, the Netherlands;
| | - Carien Bus-Spoor
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Nikki van Driel
- Department of Immunobiology, Biomedical Primate Research Centre, 2280 GH Rijswijk, the Netherlands
| | - Marissa L Dubbelaar
- Section Medical Physiology, Department of Neuroscience, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Corien Grit
- Section Medical Physiology, Department of Neuroscience, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Susanne M Kooistra
- Section Medical Physiology, Department of Neuroscience, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands.,MS Centrum Noord Nederland, 9722 NN Groningen, the Netherlands
| | - Zahra C Fagrouch
- Department of Virology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, the Netherlands; and
| | - Ernst J Verschoor
- Department of Virology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, the Netherlands; and
| | - Jan Bauer
- Department for Neuroimmunology, Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Bart J L Eggen
- Section Medical Physiology, Department of Neuroscience, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands.,MS Centrum Noord Nederland, 9722 NN Groningen, the Netherlands
| | - Hermie J M Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Jon D Laman
- Section Medical Physiology, Department of Neuroscience, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands.,MS Centrum Noord Nederland, 9722 NN Groningen, the Netherlands
| | - Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, 2280 GH Rijswijk, the Netherlands.,Section Medical Physiology, Department of Neuroscience, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands.,MS Centrum Noord Nederland, 9722 NN Groningen, the Netherlands
| |
Collapse
|
12
|
Yan L, Jiang B, Niu Y, Wang H, Li E, Yan Y, Sun H, Duan Y, Chang S, Chen G, Ji W, Xu RH, Si W. Intrathecal delivery of human ESC-derived mesenchymal stem cell spheres promotes recovery of a primate multiple sclerosis model. Cell Death Discov 2018; 4:28. [PMID: 30131877 PMCID: PMC6102276 DOI: 10.1038/s41420-018-0091-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/27/2018] [Accepted: 07/05/2018] [Indexed: 12/31/2022] Open
Abstract
Nonhuman primate experimental autoimmune encephalomyelitis (EAE) is a valuable model for multiple sclerosis, an inflammatory demyelinating disease in the central nervous system (CNS). Human embryonic stem cell-derived mesenchymal stem cells (EMSC) are effective in treating murine EAE. Yet, it remains unknown whether the EMSC efficacy is translatable to humans. Here we induced a primate EAE model in cynomolgus monkeys and delivered EMSC in spheres (EMSCsp) to preserve the cell viability during long-distance transportation. EMSCsp intrathecally injected into the CNS, remarkably reduced the clinical symptoms, brain lesions, and neuronal demyelination in the EAE monkeys during a 3-month observation. Whereas, symptoms in the vehicle control-injected EAE monkey remained and reduced slowly and MRI lesions in brain expanded. Moreover, EMSC could transdifferentiate into neural cells in vivo in the CNS of the treated animals. Supporting evidence demonstrated that EMSCsp cells cultured in cerebrospinal fluid from the EAE monkeys largely converted to neural cells with elevated expression of genes for neuronal markers, neurotrophic factors, and neuronal myelination. Thus, this study demonstrates that EMSCsp injected directly into the CNS, can attenuate the disease progression in the primate EAE model, highly encouraging for clinical translation.
Collapse
Affiliation(s)
- Li Yan
- Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Bin Jiang
- Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Yuyu Niu
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan China
| | - Hongxuan Wang
- Faculty of Health Sciences, University of Macau, Taipa, Macau China
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Enqin Li
- Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Yaping Yan
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan China
| | - Huiyan Sun
- Faculty of Health Sciences, University of Macau, Taipa, Macau China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, Jilin China
| | - Yanchao Duan
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan China
| | - Shaohui Chang
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan China
| | - Guokai Chen
- Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Weizhi Ji
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan China
| | - Ren-He Xu
- Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Wei Si
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan China
| |
Collapse
|
13
|
Kametani Y, Shiina T, Suzuki R, Sasaki E, Habu S. Comparative immunity of antigen recognition, differentiation, and other functional molecules: similarities and differences among common marmosets, humans, and mice. Exp Anim 2018; 67:301-312. [PMID: 29415910 PMCID: PMC6083031 DOI: 10.1538/expanim.17-0150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The common marmoset (CM; Callithrix jacchus) is a small New World monkey
with a high rate of pregnancy and is maintained in closed colonies as an experimental
animal species. Although CMs are used for immunological research, such as studies of
autoimmune disease and infectious disease, their immunological characteristics are less
defined than those of other nonhuman primates. We and others have analyzed antigen
recognition-related molecules, the development of hematopoietic stem cells (HSCs), and the
molecules involved in the immune response. CMs systemically express Caja-G, a major
histocompatibility complex class I molecule, and the ortholog of HLA-G, a suppressive
nonclassical HLA class I molecule. HSCs express CD117, while CD34 is not essential for
multipotency. CD117+ cells developed into all hematopoietic cell lineages, but compared
with human HSCs, B cells did not extensively develop when HSCs were transplanted into an
immunodeficient mouse. Although autoimmune models have been successfully established,
sensitization of CMs with some bacteria induced a low protective immunity. In CMs, B cells
were observed in the periphery, but IgG levels were very low compared with those in humans
and mice. This evidence suggests that CM immunity is partially suppressed systemically.
Such immune regulation might benefit pregnancy in CMs, which normally deliver dizygotic
twins, the placentae of which are fused and the immune cells of which are mixed. In this
review, we describe the CM immune system and discuss the possibility of using CMs as a
model of human immunity.
Collapse
Affiliation(s)
- Yoshie Kametani
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara-shi, Kanagawa 259-1193, Japan
| | - Takashi Shiina
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara-shi, Kanagawa 259-1193, Japan
| | - Ryuji Suzuki
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, 18-1 Sakuradai, Minami-ku, Sagamihara-shi, Kanagawa 252-0392, Japan
| | - Erika Sasaki
- Central Institute for Experimental Animals,3-25-12 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-0821, Japan
| | - Sonoko Habu
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
14
|
't Hart BA, Laman JD, Kap YS. Reverse Translation for Assessment of Confidence in Animal Models of Multiple Sclerosis for Drug Discovery. Clin Pharmacol Ther 2017; 103:262-270. [DOI: 10.1002/cpt.801] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/06/2017] [Accepted: 07/17/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Bert A. 't Hart
- Department Immunobiology; Biomedical Primate Research Centre; Rijswijk The Netherlands
- University of Groningen, University Medical Centre, Dept. Neuroscience; Groningen The Netherlands
- MS Center Noord-Nederland; Groningen The Netherlands
| | - Jon D. Laman
- University of Groningen, University Medical Centre, Dept. Neuroscience; Groningen The Netherlands
- MS Center Noord-Nederland; Groningen The Netherlands
| | - Yolanda S. Kap
- Department Immunobiology; Biomedical Primate Research Centre; Rijswijk The Netherlands
| |
Collapse
|
15
|
't Hart BA, Dunham J, Faber BW, Laman JD, van Horssen J, Bauer J, Kap YS. A B Cell-Driven Autoimmune Pathway Leading to Pathological Hallmarks of Progressive Multiple Sclerosis in the Marmoset Experimental Autoimmune Encephalomyelitis Model. Front Immunol 2017; 8:804. [PMID: 28744286 PMCID: PMC5504154 DOI: 10.3389/fimmu.2017.00804] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 06/26/2017] [Indexed: 12/20/2022] Open
Abstract
The absence of pathological hallmarks of progressive multiple sclerosis (MS) in commonly used rodent models of experimental autoimmune encephalomyelitis (EAE) hinders the development of adequate treatments for progressive disease. Work reviewed here shows that such hallmarks are present in the EAE model in marmoset monkeys (Callithrix jacchus). The minimal requirement for induction of progressive MS pathology is immunization with a synthetic peptide representing residues 34–56 from human myelin oligodendrocyte glycoprotein (MOG) formulated with a mineral oil [incomplete Freund’s adjuvant (IFA)]. Pathological aspects include demyelination of cortical gray matter with microglia activation, oxidative stress, and redistribution of iron. When the peptide is formulated in complete Freund’s adjuvant, which contains mycobacteria that relay strong activation signals to myeloid cells, oxidative damage pathways are strongly boosted leading to more intensive pathology. The proven absence of immune potentiating danger signals in the MOG34–56/IFA formulation implies that a narrow population of antigen-experienced T cells present in the monkey’s immune repertoire is activated. This novel pathway involves the interplay of lymphocryptovirus-infected B cells with MHC class Ib/Caja-E restricted CD8+ CD56+ cytotoxic T lymphocytes.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, Netherlands.,Department of Neuroscience, University of Groningen, University Medical Center, Groningen, Netherlands
| | - Jordon Dunham
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, Netherlands.,Department of Neuroscience, University of Groningen, University Medical Center, Groningen, Netherlands
| | - Bart W Faber
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| | - Jon D Laman
- Department of Neuroscience, University of Groningen, University Medical Center, Groningen, Netherlands.,MS Center Noord-Nederland, Groningen, Netherlands
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Jan Bauer
- Department of Neuroimmunology, Brain Research Institute, Medical University Vienna, Vienna, Austria
| | - Yolanda S Kap
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, Netherlands
| |
Collapse
|
16
|
Morandi E, Jagessar SA, 't Hart BA, Gran B. EBV Infection Empowers Human B Cells for Autoimmunity: Role of Autophagy and Relevance to Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2017; 199:435-448. [PMID: 28592428 DOI: 10.4049/jimmunol.1700178] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/05/2017] [Indexed: 11/19/2022]
Abstract
The efficacy of B cell depletion therapy in multiple sclerosis indicates their central pathogenic role in disease pathogenesis. The B lymphotropic EBV is a major risk factor in multiple sclerosis, via as yet unclear mechanisms. We reported in a nonhuman primate experimental autoimmune encephalomyelitis model that an EBV-related lymphocryptovirus enables B cells to protect a proteolysis-sensitive immunodominant myelin oligodendrocyte glycoprotein (MOG) epitope (residues 40-48) against destructive processing. This facilitates its cross-presentation to autoaggressive cytotoxic MHC-E-restricted CD8+CD56+ T cells. The present study extends these observations to intact human B cells and identifies a key role of autophagy. EBV infection upregulated APC-related markers on B cells and activated the cross-presentation machinery. Although human MOG protein was degraded less in EBV-infected than in uninfected B cells, induction of cathepsin G activity by EBV led to total degradation of the immunodominant peptides MOG35-55 and MOG1-20 Inhibition of cathepsin G or citrullination of the arginine residue within an LC3-interacting region motif of immunodominant MOG peptides abrogated their degradation. Internalized MOG colocalized with autophagosomes, which can protect from destructive processing. In conclusion, EBV infection switches MOG processing in B cells from destructive to productive and facilitates cross-presentation of disease-relevant epitopes to CD8+ T cells.
Collapse
Affiliation(s)
- Elena Morandi
- Division of Clinical Neuroscience, University of Nottingham School of Medicine, Nottingham NG7 2UH, United Kingdom
| | - S Anwar Jagessar
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk 2288, the Netherlands
| | - Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk 2288, the Netherlands.,Department of Neuroscience, University Medical Center, University of Groningen, Groningen 9700, the Netherlands; and
| | - Bruno Gran
- Division of Clinical Neuroscience, University of Nottingham School of Medicine, Nottingham NG7 2UH, United Kingdom; .,Department of Neurology, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
17
|
Stimmer L, Fovet CM, Serguera C. Experimental Models of Autoimmune Demyelinating Diseases in Nonhuman Primates. Vet Pathol 2017; 55:27-41. [DOI: 10.1177/0300985817712794] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human idiopathic inflammatory demyelinating diseases (IIDD) are a heterogeneous group of autoimmune inflammatory and demyelinating disorders of the central nervous system (CNS). These include multiple sclerosis (MS), the most common chronic IIDD, but also rarer disorders such as acute disseminated encephalomyelitis (ADEM) and neuromyelitis optica (NMO). Great efforts have been made to understand the pathophysiology of MS, leading to the development of a few effective treatments. Nonetheless, IIDD still require a better understanding of the causes and underlying mechanisms to implement more effective therapies and diagnostic methods. Experimental autoimmune encephalomyelitis (EAE) is a commonly used animal model to study the pathophysiology of IIDD. EAE is principally induced through immunization with myelin antigens combined with immune-activating adjuvants. Nonhuman primates (NHP), the phylogenetically closest relatives of humans, challenged by similar microorganisms as other primates may recapitulate comparable immune responses to that of humans. In this review, the authors describe EAE models in 3 NHP species: rhesus macaques ( Macaca mulatta), cynomolgus macaques ( Macaca fascicularis), and common marmosets ( Callithrix jacchus), evaluating their respective contribution to the understanding of human IIDD. EAE in NHP is a heterogeneous disease, including acute monophasic and chronic polyphasic forms. This diversity makes it a versatile model to use in translational research. This clinical variability also creates an opportunity to explore multiple facets of immune-mediated mechanisms of neuro-inflammation and demyelination as well as intrinsic protective mechanisms. Here, the authors review current insights into the pathogenesis and immunopathological mechanisms implicated in the development of EAE in NHP.
Collapse
Affiliation(s)
- Lev Stimmer
- U1169/US27 Platform for experimental pathology, Molecular Imaging Research Center, INSERM-CEA, Fontenay-aux-Roses, France
| | - Claire-Maëlle Fovet
- U1169/US27 Platform for general surgery, Molecular Imaging Research Center, INSERM-CEA, Fontenay-aux-Roses, France
| | - Ché Serguera
- US27, Molecular Imaging Research Center, INSERM-CEA, Fontenay-aux-Roses, France
| |
Collapse
|
18
|
Peschl P, Bradl M, Höftberger R, Berger T, Reindl M. Myelin Oligodendrocyte Glycoprotein: Deciphering a Target in Inflammatory Demyelinating Diseases. Front Immunol 2017; 8:529. [PMID: 28533781 PMCID: PMC5420591 DOI: 10.3389/fimmu.2017.00529] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/19/2017] [Indexed: 12/23/2022] Open
Abstract
Myelin oligodendrocyte glycoprotein (MOG), a member of the immunoglobulin (Ig) superfamily, is a myelin protein solely expressed at the outermost surface of myelin sheaths and oligodendrocyte membranes. This makes MOG a potential target of cellular and humoral immune responses in inflammatory demyelinating diseases. Due to its late postnatal developmental expression, MOG is an important marker for oligodendrocyte maturation. Discovered about 30 years ago, it is one of the best-studied autoantigens for experimental autoimmune models for multiple sclerosis (MS). Human studies, however, have yielded controversial results on the role of MOG, especially MOG antibodies (Abs), as a biomarker in MS. But with improved detection methods using different expression systems to detect Abs in patients' samples, this is meanwhile no longer the case. Using cell-based assays with recombinant full-length, conformationally intact MOG, several recent studies have revealed that MOG Abs can be found in a subset of predominantly pediatric patients with acute disseminated encephalomyelitis (ADEM), aquaporin-4 (AQP4) seronegative neuromyelitis optica spectrum disorders (NMOSD), monophasic or recurrent isolated optic neuritis (ON), or transverse myelitis, in atypical MS and in N-methyl-d-aspartate receptor-encephalitis with overlapping demyelinating syndromes. Whereas MOG Abs are only transiently observed in monophasic diseases such as ADEM and their decline is associated with a favorable outcome, they are persistent in multiphasic ADEM, NMOSD, recurrent ON, or myelitis. Due to distinct clinical features within these diseases it is controversially disputed to classify MOG Ab-positive cases as a new disease entity. Neuropathologically, the presence of MOG Abs is characterized by MS-typical demyelination and oligodendrocyte pathology associated with Abs and complement. However, it remains unclear whether MOG Abs are a mere inflammatory bystander effect or truly pathogenetic. This article provides deeper insight into recent developments, the clinical relevance of MOG Abs and their role in the immunpathogenesis of inflammatory demyelinating disorders.
Collapse
Affiliation(s)
- Patrick Peschl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Romana Höftberger
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
19
|
Dunham J, van Driel N, Eggen BJ, Paul C, 't Hart BA, Laman JD, Kap YS. Analysis of the cross-talk of Epstein-Barr virus-infected B cells with T cells in the marmoset. Clin Transl Immunology 2017; 6:e127. [PMID: 28243437 PMCID: PMC5311918 DOI: 10.1038/cti.2017.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 02/06/2023] Open
Abstract
Despite the well-known association of Epstein–Barr virus (EBV), a lymphocryptovirus (LCV), with multiple sclerosis, a clear pathogenic role for disease progression has not been established. The translationally relevant experimental autoimmune encephalomyelitis (EAE) model in marmoset monkeys revealed that LCV-infected B cells have a central pathogenic role in the activation of T cells that drive EAE progression. We hypothesized that LCV-infected B cells induce T-cell functions relevant for EAE progression. In the current study, we examined the ex vivo cross-talk between lymph node mononuclear cells (MNCs) from EAE marmosets and (semi-) autologous EBV-infected B-lymphoblastoid cell lines (B-LCLs). Results presented here demonstrate that infection with EBV B95-8 has a strong impact on gene expression profile of marmoset B cells, particularly those involved with antigen processing and presentation or co-stimulation to T cells. At the cellular level, we observed that MNC co-culture with B-LCLs induced decrease of CCR7 expression on T cells from EAE responder marmosets, but not in EAE monkeys without clinically evident disease. B-LCL interaction with T cells also resulted in significant loss of CD27 expression and reduced expression of IL-23R and CCR6, which coincided with enhanced IL-17A production. These results highlight the profound impact that EBV-infected B-LCL cells can have on second and third co-stimulatory signals involved in (autoreactive) T-cell activation.
Collapse
Affiliation(s)
- Jordon Dunham
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands; Department of Neuroscience, University Groningen, University Medical Center, Groningen, The Netherlands
| | - Nikki van Driel
- Department of Immunobiology, Biomedical Primate Research Centre , Rijswijk, The Netherlands
| | - Bart Jl Eggen
- Department of Neuroscience, University Groningen, University Medical Center , Groningen, The Netherlands
| | - Chaitali Paul
- Department of Neuroscience, University Groningen, University Medical Center , Groningen, The Netherlands
| | - Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands; Department of Neuroscience, University Groningen, University Medical Center, Groningen, The Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Groningen, University Medical Center , Groningen, The Netherlands
| | - Yolanda S Kap
- Department of Immunobiology, Biomedical Primate Research Centre , Rijswijk, The Netherlands
| |
Collapse
|
20
|
't Hart BA, Kap YS. An essential role of virus-infected B cells in the marmoset experimental autoimmune encephalomyelitis model. Mult Scler J Exp Transl Clin 2017; 3:2055217317690184. [PMID: 28607749 PMCID: PMC5466146 DOI: 10.1177/2055217317690184] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 12/28/2016] [Indexed: 12/16/2022] Open
Abstract
Infection with Epstein–Barr virus (EBV) has been associated with an enhanced risk of genetically susceptible individuals to develop multiple sclerosis (MS). However, an explanation for the contrast between the high EBV infection prevalence (60–90%) and the low MS prevalence (0.1%) eludes us. Here we propose a new concept for the EBV–MS association developed in the experimental autoimmune encephalomyelitis model in marmoset monkeys, which are naturally infected with the EBV-related γ1-herpesvirus CalHV3. The data indicate that the infection of B cells with a γ1-herpesvirus endows them with the capacity to activate auto-aggressive CD8+ T cells specific for myelin oligodendrocyte glycoprotein.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Yolanda S Kap
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| |
Collapse
|
21
|
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
|
22
|
'tHart BA, Kap YS, Morandi E, Laman JD, Gran B. EBV Infection and Multiple Sclerosis: Lessons from a Marmoset Model. Trends Mol Med 2016; 22:1012-1024. [PMID: 27836419 DOI: 10.1016/j.molmed.2016.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/11/2016] [Accepted: 10/13/2016] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis (MS) is thought to be initiated by the interaction of genetic and environmental factors, eliciting an autoimmune attack on the central nervous system. Epstein-Barr virus (EBV) is the strongest infectious risk factor, but an explanation for the paradox between high infection prevalence and low MS incidence remains elusive. We discuss new data using marmosets with experimental autoimmune encephalomyelitis (EAE) - a valid primate model of MS. The findings may help to explain how a common infection can contribute to the pathogenesis of MS. We propose that EBV infection induces citrullination of peptides in conjunction with autophagy during antigen processing, endowing B cells with the capacity to cross-present autoantigen to CD8+CD56+ T cells, thereby leading to MS progression.
Collapse
Affiliation(s)
- Bert A 'tHart
- Department of Immunobiology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands; University of Groningen, University Medical Center, Department of Neuroscience, Groningen, The Netherlands.
| | - Yolanda S Kap
- Department of Immunobiology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Elena Morandi
- Division of Clinical Neuroscience, University of Nottingham School of Medicine, Nottingham, UK
| | - Jon D Laman
- University of Groningen, University Medical Center, Department of Neuroscience, Groningen, The Netherlands
| | - Bruno Gran
- Division of Clinical Neuroscience, University of Nottingham School of Medicine, Nottingham, UK; Department of Neurology, Nottingham University Hospitals National Health Service (NHS) Trust, Nottingham, UK
| |
Collapse
|
23
|
Jagessar SA, Holtman IR, Hofman S, Morandi E, Heijmans N, Laman JD, Gran B, Faber BW, van Kasteren SI, Eggen BJL, 't Hart BA. Lymphocryptovirus Infection of Nonhuman Primate B Cells Converts Destructive into Productive Processing of the Pathogenic CD8 T Cell Epitope in Myelin Oligodendrocyte Glycoprotein. THE JOURNAL OF IMMUNOLOGY 2016; 197:1074-88. [PMID: 27412414 DOI: 10.4049/jimmunol.1600124] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/06/2016] [Indexed: 12/27/2022]
Abstract
EBV is the major infectious environmental risk factor for multiple sclerosis (MS), but the underlying mechanisms remain obscure. Patient studies do not allow manipulation in vivo. We used the experimental autoimmune encephalomyelitis (EAE) models in the common marmoset and rhesus monkey to model the association of EBV and MS. We report that B cells infected with EBV-related lymphocryptovirus (LCV) are requisite APCs for MHC-E-restricted autoaggressive effector memory CTLs specific for the immunodominant epitope 40-48 of myelin oligodendrocyte glycoprotein (MOG). These T cells drive the EAE pathogenesis to irreversible neurologic deficit. The aim of this study was to determine why LCV infection is important for this pathogenic role of B cells. Transcriptome comparison of LCV-infected B cells and CD20(+) spleen cells from rhesus monkeys shows increased expression of genes encoding elements of the Ag cross-presentation machinery (i.e., of proteasome maturation protein and immunoproteasome subunits) and enhanced expression of MHC-E and of costimulatory molecules (CD70 and CD80, but not CD86). It was also shown that altered expression of endolysosomal proteases (cathepsins) mitigates the fast endolysosomal degradation of the MOG40-48 core epitope. Finally, LCV infection also induced expression of LC3-II(+) cytosolic structures resembling autophagosomes, which seem to form an intracellular compartment where the MOG40-48 epitope is protected against proteolytic degradation by the endolysosomal serine protease cathepsin G. In conclusion, LCV infection induces a variety of changes in B cells that underlies the conversion of destructive processing of the immunodominant MOG40-48 epitope into productive processing and cross-presentation to strongly autoaggressive CTLs.
Collapse
Affiliation(s)
- S Anwar Jagessar
- Department of Immunobiology, Biomedical Primate Research Centre, 2288GJ Rijswijk, the Netherlands; Department of Immunology, Erasmus University Medical Center, 3015CE Rotterdam, the Netherlands; MS Centre ErasMS, 3015CE Rotterdam, the Netherlands
| | - Inge R Holtman
- Department of Neuroscience, University Medical Center, University Groningen, 9713AV Groningen, the Netherlands
| | - Sam Hofman
- Department of Immunobiology, Biomedical Primate Research Centre, 2288GJ Rijswijk, the Netherlands
| | - Elena Morandi
- Division of Clinical Neuroscience, University of Nottingham School of Medicine, NG7 2UH Nottingham, United Kingdom
| | - Nicole Heijmans
- Department of Immunobiology, Biomedical Primate Research Centre, 2288GJ Rijswijk, the Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Medical Center, University Groningen, 9713AV Groningen, the Netherlands
| | - Bruno Gran
- Division of Clinical Neuroscience, University of Nottingham School of Medicine, NG7 2UH Nottingham, United Kingdom
| | - Bart W Faber
- Department of Parasitology, Biomedical Primate Research Centre, 2288GJ Rijswijk, the Netherlands; and
| | - Sander I van Kasteren
- Leiden Institute of Chemistry and The Institute for Chemical Immunology, Leiden University, 2333CC Leiden, the Netherlands
| | - Bart J L Eggen
- Department of Neuroscience, University Medical Center, University Groningen, 9713AV Groningen, the Netherlands
| | - Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, 2288GJ Rijswijk, the Netherlands; Department of Immunology, Erasmus University Medical Center, 3015CE Rotterdam, the Netherlands; Department of Neuroscience, University Medical Center, University Groningen, 9713AV Groningen, the Netherlands;
| |
Collapse
|
24
|
Kap YS, Jagessar SA, Dunham J, 't Hart BA. The common marmoset as an indispensable animal model for immunotherapy development in multiple sclerosis. Drug Discov Today 2016; 21:1200-5. [PMID: 27060373 DOI: 10.1016/j.drudis.2016.03.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/16/2016] [Accepted: 03/31/2016] [Indexed: 12/21/2022]
Abstract
New drugs often fail in the translation from the rodent experimental autoimmune encephalomyelitis (EAE) model to human multiple sclerosis (MS). Here, we present the marmoset EAE model as an indispensable model for translational research into MS. The genetic heterogeneity of this species and lifelong exposure to chronic latent infections and environmental pathogens create a human-like immune system. Unique to this model is the presence of the pathological hallmark of progressive MS, in particular cortical grey matter lesions. Another great possibility of this model is systemic and longitudinal immune profiling, whereas in humans and mice immune profiling is usually performed in a single compartment (i.e. blood or spleen, respectively). Overall, the marmoset model provides unique opportunities for systemic drug-effect profiling.
Collapse
Affiliation(s)
- Yolanda S Kap
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | - S Anwar Jagessar
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Jordon Dunham
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands; Department of Medical Physiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands; Department of Medical Physiology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
25
|
Luo H, Broux B, Wang X, Hu Y, Ghannam S, Jin W, Larochelle C, Prat A, Wu J. EphrinB1 and EphrinB2 regulate T cell chemotaxis and migration in experimental autoimmune encephalomyelitis and multiple sclerosis. Neurobiol Dis 2016; 91:292-306. [PMID: 27039370 DOI: 10.1016/j.nbd.2016.03.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 03/14/2016] [Accepted: 03/16/2016] [Indexed: 12/27/2022] Open
Abstract
T cells are believed to be key effector cells in multiple sclerosis (MS). In this study, we examined the roles of T cell ephrinB1 (EFNB1) and ephrinB2 (EFNB2) in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and MS. We provide evidence that animals with T cell specific double deletion of EFNB1 and EFNB2 (dKO) have reduced proliferation in response to MOG35-55, defective Th1 and Th17 differentiations and significantly lower scores of MOG-induced EAE. We further demonstrate that dKO T cells are compromised in their ability to migrate into the CNS of EAE animals in vivo and towards multiple chemokines in vitro. Using deletion mutations, we identified a critical 11-aa EFNB1 intracellular domain segment that controls T cell chemotaxis towards CCL21. In humans, EFNB1 and EFNB2 are highly expressed in Th1 and Th17 cells and EFNB1- and EFNB2-expressing T cells are found among immune cell infiltrates in MS lesions. Reverse signaling through EFNB1 and EFNB2 in human Th17 cells enhances their migration through a monolayer of blood brain barrier endothelial cells. Our study demonstrates that expression of EFNB1 and EFNB2 is implicated in Th cell differentiation and migration to inflammatory sites in both EAE and MS.
Collapse
Affiliation(s)
- Hongyu Luo
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Bieke Broux
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Department of Neuroscience, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Hasselt University, Biomedical Research Institute and transnationale Universiteit Limburg, School of Life Sciences, Agoralaan building C, 3590 Diepenbeek, Belgium
| | - Xuehai Wang
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Yan Hu
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Soufiane Ghannam
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Department of Neuroscience, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Wei Jin
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Catherine Larochelle
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Department of Neuroscience, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada
| | - Alexandre Prat
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Department of Neuroscience, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada.
| | - Jiangping Wu
- The Research Center of the University of Montreal Hospital Center e (CRCHUM), 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada; Division of Nephrology, Department of Medicine, Faculty of Medicine, Université de Montréal, 900 Rue Saint-Denis, Montréal H2X 0A9, Québec, Canada.
| |
Collapse
|
26
|
'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
|
27
|
Jagessar SA, Heijmans N, Blezer ELA, Bauer J, Weissert R, 't Hart BA. Immune profile of an atypical EAE model in marmoset monkeys immunized with recombinant human myelin oligodendrocyte glycoprotein in incomplete Freund's adjuvant. J Neuroinflammation 2015; 12:169. [PMID: 26377397 PMCID: PMC4574133 DOI: 10.1186/s12974-015-0378-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/16/2015] [Indexed: 11/21/2022] Open
Abstract
Background Experimental autoimmune encephalomyelitis (EAE) in the common marmoset monkey (Callithrix jacchus) is a relevant preclinical model for translational research into immunopathogenic mechanisms operating in multiple sclerosis (MS). Prior studies showed a core pathogenic role of T and B cells specific for myelin oligodendrocyte glycoprotein (MOG). However, in those studies, the quality of the response against MOG epitopes was strongly biased by bacterial antigens in the complete Freund’s adjuvant (CFA), in which the immunizing recombinant human (rh) MOG protein had been formulated. In response to the need of a more refined EAE model, we have tested whether disease could also be induced with rhMOG in incomplete Freund’s adjuvant (IFA). Method Marmosets were immunized with rhMOG emulsified in IFA in the dorsal skin. Monkeys that did not develop neurological deficit were given booster immunizations at 28-day interval with the same antigen preparation. In a second experiment, three marmoset twin pairs were sensitized against MOG peptides in IFA to study a possibility for suppressive activity towards pathogenic T cells directed against the encephalitogenic epitope MOG40-48. Results Despite the absence of strong danger signals in the rhMOG/IFA inoculum, all monkeys developed clinically evident EAE symptoms. Moreover, in all monkeys, demyelinated lesions were present in the white matter and in two cases also in the cortical grey matter. Immune profiling at height of the disease showed a dominant T cell response against the overlapping peptides 14–36 and 24–46, but reactivity against the pathogenically most relevant peptide 34–56 was conspicuously absent. In the second experiment, there was an indication for a possible suppressive mechanism. Conclusions Immunization of marmoset monkeys with rhMOG in IFA elicits clinical EAE in all animals. Moreover, rhMOG contains pathogenic and regulatory epitopes, but the pathogenic hierarchy of rhMOG epitopes is strongly influenced by the adjuvant in which the protein is formulated. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0378-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- S Anwar Jagessar
- Department of Immunobiology, Biomedical Primate Research Centre, P.O. Box 3306, 2280, GH, Rijswijk, The Netherlands. .,ErasMS Centre, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Nicole Heijmans
- Department of Immunobiology, Biomedical Primate Research Centre, P.O. Box 3306, 2280, GH, Rijswijk, The Netherlands
| | - Erwin L A Blezer
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Bauer
- Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| | - Robert Weissert
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, P.O. Box 3306, 2280, GH, Rijswijk, The Netherlands. .,ErasMS Centre, Erasmus Medical Center, Rotterdam, The Netherlands. .,Department of Neuroscience, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
28
|
׳t Hart BA. Reverse translation of failed treatments can help improving the validity of preclinical animal models. Eur J Pharmacol 2015; 759:14-8. [DOI: 10.1016/j.ejphar.2015.03.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/16/2015] [Accepted: 03/12/2015] [Indexed: 10/23/2022]
|
29
|
Kap YS, van Driel N, Arends R, Rouwendal G, Verolin M, Blezer E, Lycke N, 't Hart BA. Immune modulation by a tolerogenic myelin oligodendrocyte glycoprotein (MOG)10-60 containing fusion protein in the marmoset experimental autoimmune encephalomyelitis model. Clin Exp Immunol 2015; 180:28-39. [PMID: 25393803 DOI: 10.1111/cei.12487] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2014] [Indexed: 01/11/2023] Open
Abstract
Current therapies for multiple sclerosis (MS), a chronic autoimmune neuroinflammatory disease, mostly target general cell populations or immune molecules, which may lead to a compromised immune system. A more directed strategy would be to re-enforce tolerance of the autoaggressive T cells that drive tissue inflammation and injury. In this study, we have investigated whether the course of experimental autoimmune encephalomyelitis (EAE) in mice and marmosets can be altered by a potent tolerizing fusion protein. In addition, a multi-parameter immunological analysis was performed in marmosets to assess whether the treatment induces modulation of EAE-associated cellular and humoral immune reactions. The fusion protein, CTA1R9K-hMOG10-60-DD, contains a mutated cholera toxin A1 subunit (CTA1R9K), a dimer of the Ig binding D region of Staphylococcus aureus protein A (DD), and the human myelin oligodendrocyte glycoprotein (hMOG) sequence 10-60. We observed that intranasal application of CTA1R9K-hMOG10-60-DD seems to skew the immune response against myelin oligodendrocyte glycoprotein (MOG) towards a regulatory function. We show a reduced number of circulating macrophages, reduced MOG-induced expansion of mononuclear cells in peripheral blood, reduced MOG-induced production of interleukin (IL)-17A in spleen, increased MOG-induced production of IL-4 and IL-10 and an increased percentage of cells expressing programmed cell death-1 (PD-1) and CC chemokine receptor 4 (CCR4). Nevertheless, the treatment did not detectably change the EAE course and pathology. Thus, despite a detectable effect on relevant immune parameters, the fusion protein failed to influence the clinical and pathological outcome of disease. This result warrants further development and improvement of this specifically targeted tolerance inducing therapy.
Collapse
Affiliation(s)
- Y S Kap
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands; MS Centre ErasMS, Rotterdam, The Netherlands; Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
30
|
't Hart BA, Bogers WM, Haanstra KG, Verreck FA, Kocken CH. The translational value of non-human primates in preclinical research on infection and immunopathology. Eur J Pharmacol 2015; 759:69-83. [PMID: 25814254 DOI: 10.1016/j.ejphar.2015.03.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/09/2015] [Accepted: 03/12/2015] [Indexed: 01/01/2023]
Abstract
The immune system plays a central role in the defense against environmental threats - such as infection with viruses, parasites or bacteria - but can also be a cause of disease, such as in the case of allergic or autoimmune disorders. In the past decades the impressive development of biotechnology has provided scientists with biological tools for the development of highly selective treatments for the different types of disorders. However, despite some clear successes the translation of scientific discoveries into effective treatments has remained challenging. The often-disappointing predictive validity of the preclinical animal models that are used in the selection of the most promising vaccine or drug candidates is the Achilles heel in the therapy development process. This publication summarizes the relevance and usage of non-human primates as pre-clinical model in infectious and autoimmune diseases, in particular for biologicals, which due to their high species-specificity are inactive in lower species.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands; University of Groningen, University Medical Center, Department Neuroscience, Groningen, The Netherlands.
| | - Willy M Bogers
- Department Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | - Krista G Haanstra
- Department Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | - Frank A Verreck
- Department Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | - Clemens H Kocken
- Department Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| |
Collapse
|
31
|
't Hart BA, van Kooyk Y, Geurts JJG, Gran B. The primate autoimmune encephalomyelitis model; a bridge between mouse and man. Ann Clin Transl Neurol 2015; 2:581-93. [PMID: 26000330 PMCID: PMC4435712 DOI: 10.1002/acn3.194] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/19/2015] [Accepted: 02/19/2015] [Indexed: 12/13/2022] Open
Abstract
Introduction Multiple sclerosis (MS) is an enigmatic autoimmune-driven inflammatory/demyelinating disease of the human central nervous system (CNS), affecting brain, spinal cord, and optic nerves. The cause of the disease is not known and the number of effective treatments is limited. Despite some clear successes, translation of immunological discoveries in the mouse experimental autoimmune encephalomyelitis (EAE) model into effective therapies for MS patients has been difficult. This translation gap between MS and its elected EAE animal model reflects the phylogenetic distance between humans and their experimental counterpart, the inbred/specific pathogen free (SPF) laboratory mouse. Objective Here, we discuss that important new insights can be obtained into the mechanistic basis of the therapy paradox from the study of nonhuman primate EAE (NHP-EAE) models, the well-validated EAE model in common marmosets (Callithrix jacchus) in particular. Interpretation Data presented in this review demonstrate that due to a considerable immunological and pathological overlap with mouse EAE on one side and MS on the other, the NHP EAE model can help us bridge the translation gap.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre Rijswijk, The Netherlands ; Department Neuroscience, University Medical Center, University of Groningen Groningen, The Netherlands
| | - Yvette van Kooyk
- Department of Cell Biology and Immunology, Free University Medical Center Amsterdam, The Netherlands
| | - Jeroen J G Geurts
- Department of Anatomy and Neuroscience, Free University Medical Center Amsterdam, The Netherlands
| | - Bruno Gran
- Division of Clinical Neuroscience, University of Nottingham School of Medicine Nottingham, United Kingdom
| |
Collapse
|
32
|
Haanstra KG, Dijkman K, Bashir N, Bauer J, Mary C, Poirier N, Baker P, Crossan CL, Scobie L, 't Hart BA, Vanhove B. Selective blockade of CD28-mediated T cell costimulation protects rhesus monkeys against acute fatal experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2015; 194:1454-66. [PMID: 25589073 DOI: 10.4049/jimmunol.1402563] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Costimulatory and coinhibitory receptor-ligand pairs on T cells and APC control the immune response. We have investigated whether selective blockade of CD28-CD80/86 costimulatory interactions, which preserves the coinhibitory CTLA4-CD80/86 interactions and the function of regulatory T (Treg) cells, abrogates the induction of experimental autoimmune encephalomyelitis (EAE) in rhesus monkeys. EAE was induced by intracutaneous immunization with recombinant human myelin oligodendrocyte glycoprotein (rhMOG) in CFA on day 0. FR104 is a monovalent, PEGylated-humanized Fab' Ab fragment against human CD28, cross-reactive with rhesus monkey CD28. FR104 or placebo was administered on days 0, 7, 14, and 21. FR104 levels remained high until the end of the study (day 42). Placebo-treated animals all developed clinical EAE between days 12 and 27. FR104-treated animals did not develop clinical EAE and were sacrificed at the end of the study resulting in a significantly prolonged survival. FR104 treatment diminished T and B cell responses against rhMOG, significantly reduced CNS inflammation and prevented demyelination. The inflammatory profile in the cerebrospinal fluid and brain material was also strongly reduced. Recrudescence of latent virus was investigated in blood, spleen, and brain. No differences between groups were observed for the β-herpesvirus CMV and the polyomaviruses SV40 and SA12. Cross-sectional measurement of lymphocryptovirus, the rhesus monkey EBV, demonstrated elevated levels in the blood of FR104-treated animals. Blocking rhesus monkey CD28 with FR104 mitigated autoreactive T and B cell activation and prevented CNS pathology in the rhMOG/CFA EAE model in rhesus monkeys.
Collapse
Affiliation(s)
- Krista G Haanstra
- Biomedical Primate Research Centre, 2280 GH Rijswijk, the Netherlands;
| | - Karin Dijkman
- Biomedical Primate Research Centre, 2280 GH Rijswijk, the Netherlands
| | - Noun Bashir
- Biomedical Primate Research Centre, 2280 GH Rijswijk, the Netherlands
| | - Jan Bauer
- Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | - Paul Baker
- Glasgow Caledonian University, Glasgow G4 0BA, United Kingdom
| | | | - Linda Scobie
- Glasgow Caledonian University, Glasgow G4 0BA, United Kingdom
| | - Bert A 't Hart
- Biomedical Primate Research Centre, 2280 GH Rijswijk, the Netherlands; University of Groningen, University Medical Center, Department of Neuroscience, 9713 GZ Groningen, the Netherlands; and
| | - Bernard Vanhove
- Effimune SAS, 44035 Nantes, France; Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1064, 44093 Nantes, France
| |
Collapse
|
33
|
Vanheusden M, Stinissen P, ’t Hart BA, Hellings N. Cytomegalovirus: a culprit or protector in multiple sclerosis? Trends Mol Med 2015; 21:16-23. [DOI: 10.1016/j.molmed.2014.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/07/2014] [Accepted: 11/14/2014] [Indexed: 12/26/2022]
|
34
|
Farooq SM, Elkhatib WF, Ashour HM. The in vivo and in vitro induction of anterior chamber associated immune deviation to myelin antigens in C57BL/6 mice. Brain Behav Immun 2014; 42:118-22. [PMID: 24953428 DOI: 10.1016/j.bbi.2014.06.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 06/09/2014] [Accepted: 06/11/2014] [Indexed: 10/25/2022] Open
Abstract
Introduction of antigens into the anterior chamber (AC) of the eye generates a specific systemic form of tolerance that is termed AC-associated immune deviation (ACAID). Experimental autoimmune encephalomyelitis (EAE) is an animal model of the human CNS demyelinating diseases, including multiple sclerosis (MS) and acute disseminated encephalomyelitis. We investigated whether the encephalitogenic antigens myelin oligodendrocyte glycoprotein (MOG35-55) or myelin basic protein (MBP) induce ACAID in the EAE-prone C57BL/6 mice. We hypothesized that injection of MOG35-55/MBP induces antigen-specific tolerance whether via the AC route, the adoptive transfer of in vitro-generated MOG35-55-specific/MBP-specific ACAID antigen presenting cells (APCs), or the adoptive transfer of MOG35-55-specific/MBP-specific ACAID T regulatory cells (Tregs). ACAID is characterized by the specific impairment of delayed-type hypersensitivity (DTH) responses. Thus, DTH assays were used to test for ACAID following the AC injection of MOG35-55/MBP, or the intravenous injection of MOG35-55-specific/MBP-specific ACAID APCs. The functional local adoptive transfer (LAT) assays were used to examine the putative regulatory functions of in vitro generated MOG35-55-specific/MBP-specific Tregs. This report is the first to demonstrate the in vivo and in vitro induction of MOG35-55-specific/MBP-specific ACAID-mediated tolerance in C57BL/6 mice. These findings highlight the need for novel immunotherapeutic strategies for MS and optic neuritis.
Collapse
Affiliation(s)
- Shukkur M Farooq
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Walid F Elkhatib
- Department of Microbiology & Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Department of Pediatrics, Eastern Virginia Medical School, Norfolk, VA, USA; Department of Pharmacy Practice, School of Pharmacy, Hampton University, Hampton, VA, USA
| | - Hossam M Ashour
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA; Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
35
|
‘t Hart BA, Jagessar SA, Kap YS, Haanstra KG, Philippens IH, Serguera C, Langermans J, Vierboom M. Improvement of preclinical animal models for autoimmune-mediated disorders via reverse translation of failed therapies. Drug Discov Today 2014; 19:1394-401. [DOI: 10.1016/j.drudis.2014.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 03/10/2014] [Accepted: 03/27/2014] [Indexed: 12/17/2022]
|
36
|
Pannemans K, Broux B, Goris A, Dubois B, Broekmans T, Van Wijmeersch B, Geraghty D, Stinissen P, Hellings N. HLA-E restricted CD8+ T cell subsets are phenotypically altered in multiple sclerosis patients. Mult Scler 2014; 20:790-801. [PMID: 24144875 DOI: 10.1177/1352458513509703] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/29/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND The importance of Qa-1 restricted CD8(+) T cells in regulating autoreactive T cell responses has been demonstrated in animal models for autoimmune disorders, including multiple sclerosis (MS). OBJECTIVE We hypothesize that their human variant, HLA-E restricted CD8(+) T cells, fulfills a similar regulatory role in man and that these cells are of importance in MS. METHODS A large cohort of MS patients and healthy controls was genotyped for the two known HLA-E polymorphisms. Flow cytometry was used to determine HLA-E expression kinetics and to phenotype HLA-E restricted CD8(+) T cells. Immunohistochemistry was performed to investigate HLA-E expression in the central nervous system (CNS) of MS patients. RESULTS HLA-E is upregulated on immune cells upon in vitro activation and this upregulation is polymorphism-dependent for T and B cells. T and B cells in lesions of MS patients show enhanced HLA-E expression. Furthermore, NKG2C(+)CD8(+) T cells of MS patients have a significantly lower Foxp3 expression, while NKG2A(+)CD8(+) T cells of MS patients produce higher levels of pro-inflammatory cytokines compared to those of healthy individuals. CONCLUSION Our study indicates that the HLA-E system is altered in MS and could play a regulatory role in disease.
Collapse
Affiliation(s)
- Kim Pannemans
- Biomedical Research Institute, Hasselt University, Belgium
| | - Bieke Broux
- Biomedical Research Institute, Hasselt University, Belgium
| | - An Goris
- Laboratory for Neuroimmunology, KULeuven, Belgium
| | | | - Tom Broekmans
- Biomedical Research Institute, Hasselt University, Belgium Department of Health Care, PHL University College, Belgium
| | | | - Daniel Geraghty
- Clinical Research Division, Fred Hutchinson Cancer Research Center, USA
| | - Piet Stinissen
- Biomedical Research Institute, Hasselt University, Belgium
| | - Niels Hellings
- Biomedical Research Institute, Hasselt University, Belgium
| |
Collapse
|
37
|
Sagar D, Masih S, Schell T, Jacobson S, Comber JD, Philip R, Wigdahl B, Jain P, Khan ZK. In vivo immunogenicity of Tax(11-19) epitope in HLA-A2/DTR transgenic mice: implication for dendritic cell-based anti-HTLV-1 vaccine. Vaccine 2014; 32:3274-84. [PMID: 24739247 DOI: 10.1016/j.vaccine.2014.03.087] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/20/2014] [Accepted: 03/26/2014] [Indexed: 01/10/2023]
Abstract
Viral oncoprotein Tax plays key roles in transformation of human T-cell leukemia virus (HTLV-1)-infected T cells leading to adult T-cell leukemia (ATL), and is the key antigen recognized during HTLV-associated myelopathy (HAM). In HLA-A2+ asymptomatic carriers as well as ATL and HAM patients, Tax(11-19) epitope exhibits immunodominance. Here, we evaluate CD8 T-cell immune response against this epitope in the presence and absence of dendritic cells (DCs) given the recent encouraging observations made with Phase 1 DC-based vaccine trial for ATL. To facilitate these studies, we first generated an HLA-A2/DTR hybrid mouse strain carrying the HLA-A2.1 and CD11c-DTR genes. We then studied CD8 T-cell immune response against Tax(11-19) epitope delivered in the absence or presence of Freund's adjuvant and/or DCs. Overall results demonstrate that naturally presented Tax epitope could initiate an antigen-specific CD8T cell response in vivo but failed to do so upon DC depletion. Presence of adjuvant potentiated Tax(11-19)-specific response. Elevated serum IL-6 levels coincided with depletion of DCs whereas decreased TGF-β was associated with adjuvant use. Thus, Tax(11-19) epitope is a potential candidate for the DC-based anti-HTLV-1 vaccine and the newly hybrid mouse strain could be used for investigating DC involvement in human class-I-restricted immune responses.
Collapse
Affiliation(s)
- Divya Sagar
- Department of Microbiology and Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Shet Masih
- Department of Microbiology and Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Todd Schell
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Steven Jacobson
- Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Brian Wigdahl
- Department of Microbiology and Immunology, and the Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Pooja Jain
- Department of Microbiology and Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Zafar K Khan
- Department of Microbiology and Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
38
|
Kap YS, van Driel N, Laman JD, Tak PP, 't Hart BA. CD20+ B cell depletion alters T cell homing. THE JOURNAL OF IMMUNOLOGY 2014; 192:4242-53. [PMID: 24696233 DOI: 10.4049/jimmunol.1303125] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Depleting mAbs against the pan B cell marker CD20 are remarkably effective in the treatment of autoimmune-mediated inflammatory disorders, but the underlying mechanisms are poorly defined. The primary objective of this study was to find a mechanistic explanation for the remarkable clinical effect of the anti-CD20 mAbs in a representative nonhuman primate autoimmune-mediated inflammatory disorder model, experimental autoimmune encephalomyelitis (EAE) in common marmosets, allowing detailed analysis of secondary lymphoid organs (SLO). We observed that the depletion of CD20(+) B cells creates a less immunostimulatory environment in the SLO reflected by reduced expression of MHC class II, CD40, CD83, and CD80/CD86. APCs isolated from SLO of B cell-depleted EAE monkeys were also less responsive to mitogenic stimulation. The depleted B cell areas were replenished by T cells, of which the majority expressed CD127 (IL-7R) and CCR7. Such effects were not detected in EAE marmosets treated with mAb against BLyS or APRIL, where B cell depletion via withdrawal of essential survival cytokines was not associated with a marked clinical effect. We propose that at least part of the efficacy of anti-CD20 mAb therapy is attributable to the sustained CCR7 expression on T cells within SLO, limiting their release into the circulation.
Collapse
Affiliation(s)
- Yolanda S Kap
- Department of Immunobiology, Biomedical Primate Research Centre, 2280 GH Rijswijk, The Netherlands
| | | | | | | | | |
Collapse
|
39
|
Jagessar SA, Vierboom M, Blezer ELA, Bauer J, Hart BA', Kap YS. Overview of models, methods, and reagents developed for translational autoimmunity research in the common marmoset (Callithrix jacchus). Exp Anim 2014; 62:159-71. [PMID: 23903050 PMCID: PMC4160941 DOI: 10.1538/expanim.62.159] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The common marmoset (Callithrix jacchus) is a small-bodied Neotropical
primate and a useful preclinical animal model for translational research into
autoimmune-mediated inflammatory diseases (AIMID), such as rheumatoid arthritis (RA) and
multiple sclerosis (MS). The animal model for MS established in marmosets has proven their
value for exploratory research into (etio) pathogenic mechanisms and for the evaluation of
new therapies that cannot be tested in lower species because of their specificity for
humans. Effective usage of the marmoset in preclinical immunological research has been
hampered by the limited availability of blood for immunological studies and of reagents
for profiling of cellular and humoral immune reactions. In this paper, we give a concise
overview of the procedures and reagents that were developed over the years in our
laboratory in marmoset models of the above-mentioned diseases.
Collapse
Affiliation(s)
- S Anwar Jagessar
- Department of Immunobiology, Biomedical Primate Research Centre, P.O. Box 3306, 2280 GH Rijswijk, The Netherlands.
| | | | | | | | | | | |
Collapse
|
40
|
Moncla LH, Ross TM, Dinis JM, Weinfurter JT, Mortimer TD, Schultz-Darken N, Brunner K, Capuano SV, Boettcher C, Post J, Johnson M, Bloom CE, Weiler AM, Friedrich TC. A novel nonhuman primate model for influenza transmission. PLoS One 2013; 8:e78750. [PMID: 24244352 PMCID: PMC3828296 DOI: 10.1371/journal.pone.0078750] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 09/16/2013] [Indexed: 12/20/2022] Open
Abstract
Studies of influenza transmission are necessary to predict the pandemic potential of emerging influenza viruses. Currently, both ferrets and guinea pigs are used in such studies, but these species are distantly related to humans. Nonhuman primates (NHP) share a close phylogenetic relationship with humans and may provide an enhanced means to model the virological and immunological events in influenza virus transmission. Here, for the first time, it was demonstrated that a human influenza virus isolate can productively infect and be transmitted between common marmosets (Callithrix jacchus), a New World monkey species. We inoculated four marmosets with the 2009 pandemic virus A/California/07/2009 (H1N1pdm) and housed each together with a naïve cage mate. We collected bronchoalveolar lavage and nasal wash samples from all animals at regular intervals for three weeks post-inoculation to track virus replication and sequence evolution. The unadapted 2009 H1N1pdm virus replicated to high titers in all four index animals by 1 day post-infection. Infected animals seroconverted and presented human-like symptoms including sneezing, nasal discharge, labored breathing, and lung damage. Transmission occurred in one cohabitating pair. Deep sequencing detected relatively few genetic changes in H1N1pdm viruses replicating in any infected animal. Together our data suggest that human H1N1pdm viruses require little adaptation to replicate and cause disease in marmosets, and that these viruses can be transmitted between animals. Marmosets may therefore be a viable model for studying influenza virus transmission.
Collapse
Affiliation(s)
- Louise H. Moncla
- Department of Pathobiological Sciences, University of Wisconsin School of Veterinary Medicine, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
- University of Wisconsin Microbiology Doctoral Training Program, Madison, Wisconsin, United States of America
| | - Ted M. Ross
- Center for Vaccine Research, Dept. of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jorge M. Dinis
- Department of Pathobiological Sciences, University of Wisconsin School of Veterinary Medicine, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
- University of Wisconsin Microbiology Doctoral Training Program, Madison, Wisconsin, United States of America
| | - Jason T. Weinfurter
- Department of Pathobiological Sciences, University of Wisconsin School of Veterinary Medicine, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Tatum D. Mortimer
- Department of Pathobiological Sciences, University of Wisconsin School of Veterinary Medicine, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
- University of Wisconsin Microbiology Doctoral Training Program, Madison, Wisconsin, United States of America
| | - Nancy Schultz-Darken
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Kevin Brunner
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Saverio V. Capuano
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Carissa Boettcher
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Jennifer Post
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Michael Johnson
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Chalise E. Bloom
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida, United States of America
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Thomas C. Friedrich
- Department of Pathobiological Sciences, University of Wisconsin School of Veterinary Medicine, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
- University of Wisconsin Microbiology Doctoral Training Program, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
41
|
The repertoire of MHC class I genes in the common marmoset: evidence for functional plasticity. Immunogenetics 2013; 65:841-9. [PMID: 24018468 DOI: 10.1007/s00251-013-0732-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 08/24/2013] [Indexed: 12/23/2022]
Abstract
In humans, the classical antigen presentation function of major histocompatibility complex (MHC) class I molecules is controlled by the human leukocyte antigen HLA -A, HLA-B and HLA-C loci. A similar observation has been made for great apes and Old World monkey species. In contrast, a New World monkey species such as the cotton-top tamarin (Saguinus oedipus) appears to employ the G locus for its classical antigen presentation function. At present, little is known about the classical MHC class I repertoire of the common marmoset (Callithrix jacchus), another New World monkey that is widely used in biomedical research. In the present population study, no evidence has been found for abundant transcription of classical I class genes. However, in each common marmoset, four to seven different G-like alleles were detected, suggesting that the ancestral locus has been subject to expansion. Segregation studies provided evidence for at least two G-like genes present per haplotype, which are transcribed by a variety of cell types. The alleles of these Caja-G genes cluster in separate lineages, suggesting that the loci diversified considerably after duplication. Phylogenetic analyses of the introns confirm that the Caja-G loci cluster in the vicinity of HLA-G, indicating that both genes shared an ancestor. In contrast to HLA-G, Caja-G shows considerable polymorphism at the peptide-binding sites. This observation, together with the lack of detectable transcripts of A and B-like genes, indicates that Caja-G genes have taken over the function of classical class I genes. These data highlight the extreme plasticity of the MHC class I gene system.
Collapse
|
42
|
Induction of encephalitis in rhesus monkeys infused with lymphocryptovirus-infected B-cells presenting MOG(34-56) peptide. PLoS One 2013; 8:e71549. [PMID: 23977076 PMCID: PMC3744571 DOI: 10.1371/journal.pone.0071549] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 07/01/2013] [Indexed: 12/18/2022] Open
Abstract
The overlapping epidemiology of multiple sclerosis (MS) and Epstein-Barr virus (EBV), the increased risk to develop MS after infectious mononucleosis (IM) and the localization of EBV-infected B-cells within the MS brain suggest a causal link between EBV and MS. However, the underlying mechanism is unknown. We hypothesize that EBV-infected B-cells are capable of eliciting a central nervous system (CNS) targeting autoimmune reaction. To test this hypothesis we have developed a novel experimental model in rhesus monkeys of IM-like disease induced by infusing autologous B-lymphoblastoid cells (B-LCL). Herpesvirus papio (HVP) is a lymphocryptovirus related to EBV and was used to generate rhesus monkey B-LCL. Three groups of five animals were included; each group received three intravenous infusions of B-LCL that were either pulsed with the encephalitogenic self peptide MOG34–56 (group A), a mimicry peptide (981–1003) of the major capsid protein of cytomegalovirus (CMVmcp981–1003; group B) or the citrullinated MOG34–56 (cMOG34–56; group C). Groups A and B received on day 98 a single immunization with MOG34–56 in incomplete Freund’s adjuvant (IFA). Group C monkeys were euthanized just prior to day 98 without booster immunization. We observed self-peptide-specific proliferation of T-cells, superimposed on similar strong proliferation of CD3+CD8+ T-cells against the B-LCL as observed in IM. The brains of several monkeys contained perivascular inflammatory lesions of variable size, comprising CD3+ and CD68+ cells. Moreover, clusters of CD3+ and CD20+ cells were detected in the meninges. The only evident clinical sign was substantial loss of bodyweight (>15%), a symptom observed both in early autoimmune encephalitis and IM. In conclusion, this model suggests that EBV-induced B-LCL can elicit a CNS targeting inflammatory (auto)immune reaction.
Collapse
|
43
|
't Hart BA, Jagessar SA, Haanstra K, Verschoor E, Laman JD, Kap YS. The Primate EAE Model Points at EBV-Infected B Cells as a Preferential Therapy Target in Multiple Sclerosis. Front Immunol 2013; 4:145. [PMID: 23781220 PMCID: PMC3680746 DOI: 10.3389/fimmu.2013.00145] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 05/28/2013] [Indexed: 01/08/2023] Open
Abstract
The remarkable clinical efficacy of anti-CD20 monoclonal antibodies (mAb) in relapsing-remitting multiple sclerosis points at the critical involvement of B cells in the disease. However, the exact pathogenic contribution of B cells is poorly understood. In this publication we review new data on the role of CD20+ B cells in a unique experimental autoimmune encephalomyelitis (EAE) model in common marmosets (Callithrix jacchus), a small-bodied neotropical primate. We will also discuss the relevance of these data for MS. Different from rodent EAE models, but similar to MS, disease progression in marmosets can develop independent of autoantibodies. Progressive disease is mediated by MHC class Ib (Caja-E) restricted cytotoxic T cells, which are activated by γ-herpesvirus-infected B cells and cause widespread demyelination of cortical gray matter. B-cell directed monoclonal antibody therapies (anti-CD20 versus anti-BLyS and anti-APRIL) have a variable effect on EAE progression, which we found associated with variable depletion of the Epstein Barr virus (EBV)-like γ-herpesvirus CalHV3 from lymphoid organs. These findings support an important pathogenic role of CD20+ B cell in MS, especially of the subset infected with EBV.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre , Rijswijk , Netherlands ; Multiple Sclerosis Center, Erasmus MC , Rotterdam , Netherlands ; Department of Neuroscience, University Medical Center Groningen , Groningen , Netherlands ; Department of Immunology, Erasmus MC, University Medical Center Rotterdam , Rotterdam , Netherlands
| | | | | | | | | | | |
Collapse
|
44
|
Anwar Jagessar S, Fagrouch Z, Heijmans N, Bauer J, Laman JD, Oh L, Migone T, Verschoor EJ, ’t Hart BA. The Different Clinical Effects of Anti-BLyS, Anti-APRIL and Anti-CD20 Antibodies Point at a Critical Pathogenic Role of γ-Herpesvirus Infected B Cells in the Marmoset EAE Model. J Neuroimmune Pharmacol 2013; 8:727-38. [DOI: 10.1007/s11481-013-9448-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 02/27/2013] [Indexed: 11/28/2022]
|
45
|
't Hart BA, Chalan P, Koopman G, Boots AMH. Chronic autoimmune-mediated inflammation: a senescent immune response to injury. Drug Discov Today 2012. [PMID: 23195330 DOI: 10.1016/j.drudis.2012.11.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The increasing prevalence of chronic autoimmune-mediated inflammatory diseases (AIMIDs) in ageing western societies is a major challenge for the drug development industry. The current high medical need for more-effective treatments is at least in part caused by our limited understanding of the mechanisms that drive chronic inflammation. Here, we postulate a role for immunosenescence in the progression of acute to chronic inflammation via a dysregulated response to primary injury at the level of the damaged target organ. A corollary to this notion is that treatment of acute versus chronic phases of disease might require differential targeting strategies.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, P.O. Box 3306, 2280 GH Rijswijk, The Netherlands.
| | | | | | | |
Collapse
|
46
|
B-Cell Depletion Abrogates T Cell-Mediated Demyelination in an Antibody-Nondependent Common Marmoset Experimental Autoimmune Encephalomyelitis Model. J Neuropathol Exp Neurol 2012; 71:716-28. [DOI: 10.1097/nen.0b013e3182622691] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
47
|
Jagessar SA, Heijmans N, Oh L, Bauer J, Blezer ELA, Laman JD, Migone TS, Devalaraja MN, 't Hart BA. Antibodies against human BLyS and APRIL attenuate EAE development in marmoset monkeys. J Neuroimmune Pharmacol 2012; 7:557-70. [PMID: 22870852 PMCID: PMC3419352 DOI: 10.1007/s11481-012-9384-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 06/14/2012] [Indexed: 12/04/2022]
Abstract
B lymphocyte stimulator (BLyS, also indicated as BAFF (B-cell activating factor) and CD257), and A Proliferation Inducing Ligand (APRIL, CD256) are two members of the TNF superfamily with a central role in B cell survival. Antibodies against these factors have potential therapeutic relevance in autoimmune inflammatory disorders with a proven pathogenic contribution of B cells, such as multiple sclerosis (MS). In the current study we performed a multi-parameter efficacy comparison of monoclonal antibodies against human anti-BLyS and anti-APRIL in a common marmoset (Callithrix jacchus) model of experimental autoimmune encephalomyelitis (EAE). A MS-like disease was induced by immunization with recombinant human myelin/oligodendrocyte glycoprotein (rhMOG) in complete Freund’s adjuvant. The results show that the anti-BLyS and anti-APRIL antibody cause significant depletion of circulating CD20+ B cells, but a small subset of CD20 + CD40high B cells was not depleted. Induction of CD20+ B cell depletion from lymph nodes was only observed in the anti-BLyS treated monkeys. Both antibodies had a significant inhibitory effect on disease development, but all monkeys developed clinically evident EAE. Anti-BLyS treated monkeys were sacrificed with the same clinical signs as saline-treated monkeys, but nevertheless displayed significantly reduced spinal cord demyelination. This effect was not observed in the anti-APRIL treated monkeys. The two antibodies had a different effect on T cell subset activation and the profiles of ex vivo released cytokines. In conclusion, treatment with anti-BLyS and anti-APRIL delays the development of neurological disease in a relevant preclinical model of MS. The two mAbs achieve this effect via different mechanisms.
Collapse
Affiliation(s)
- S Anwar Jagessar
- Department Immunobiology, Biomedical Primate Research Centre, PO Box 3306, 2280 GH, Rijswijk, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Discrepant effects of human interferon-gamma on clinical and immunological disease parameters in a novel marmoset model for multiple sclerosis. J Neuroimmune Pharmacol 2011; 7:253-65. [PMID: 22012268 PMCID: PMC3280389 DOI: 10.1007/s11481-011-9320-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 10/02/2011] [Indexed: 11/18/2022]
Abstract
The core pathogenic process in the common marmoset model of multiple sclerosis (MS) is the activation of memory-like T cells specific for peptide 34 to 56 derived from the extracellular domain of myelin/oligodendrocyte glycoprotein (MOG34-56). Immunization with MOG34-56 in incomplete Freund’s adjuvant is a sufficient stimulus for in vivo activation of these T cells, together with the induction of MS-like disease and CNS pathology. Ex vivo functional characteristics of MOG34-56 specific T cells are specific cytolysis of peptide pulsed target cells and high IL-17A production. To indentify possible functions in this new model of T helper 1 cells, which play a central pathogenic role in MS models induced with complete Freund’s adjuvant, we tested the effect of human interferon-γ (IFNγ) administration during disease initiation of the disease (day 0–25) and around the time of disease expression (psd 56–81). The results show a clear modulatory effect of early IFNγ treatment on humoral and cellular autoimmune parameters, but no generalized mitigating effect on the disease course. These results argue against a prominent pathogenic role of T helper 1 cells in this new marmoset EAE model.
Collapse
|