1
|
Li C, Huynh NPT, Schanz SJ, Windrem MS, Goldman SA. JC virus spread is potentiated by glial replication and demyelination-linked glial proliferation. Brain 2024; 147:4131-4146. [PMID: 39133566 DOI: 10.1093/brain/awae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 06/02/2024] [Accepted: 06/29/2024] [Indexed: 11/14/2024] Open
Abstract
Progressive multifocal leukoencephalopathy is a demyelinating infection of the immunosuppressed brain, mediated by the gliotropic polyomavirus JCV. JCV replicates in human glial progenitor cells and astrocytes, which undergo viral T-antigen-triggered mitosis, enabling viral replication. We asked whether JCV spread might therefore be accelerated by glial proliferation. Using both in vitro analysis and a human glial chimeric mouse model of JCV infection, we found that dividing human astrocytes supported JCV propagation to a substantially greater degree than did mitotically quiescent cells. Accordingly, bulk and single-cell RNA-sequence analysis revealed that JCV-infected glia differentially manifested cell cycle-linked disruption of both DNA damage response and transcriptional regulatory pathways. In vivo, JCV infection of humanized glial chimeras was greatly accentuated by cuprizone-induced demyelination and its associated mobilization of glial progenitor cells. Importantly, in vivo infection triggered the death of both uninfected and infected glia, reflecting significant bystander death. Together, these data suggest that JCV propagation in progressive multifocal leukoencephalopathy might be accelerated by glial cell division. As such, the accentuated glial proliferation attending disease-associated demyelination might provide an especially favourable environment for JCV propagation, thus potentiating oligodendrocytic bystander death and further accelerating demyelination in susceptible hosts.
Collapse
Affiliation(s)
- Cui Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, China
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
| | - Nguyen P T Huynh
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen 1017, Denmark
- Sana Biotechnology, Cambridge, MA 02139, USA
| | - Steven J Schanz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
| | - Martha S Windrem
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester NY 14604, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen 1017, Denmark
- Sana Biotechnology, Cambridge, MA 02139, USA
| |
Collapse
|
2
|
Klaus T, Hieber C, Bros M, Grabbe S. Integrins in Health and Disease-Suitable Targets for Treatment? Cells 2024; 13:212. [PMID: 38334604 PMCID: PMC10854705 DOI: 10.3390/cells13030212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Integrin receptors are heterodimeric surface receptors that play multiple roles regarding cell-cell communication, signaling, and migration. The four members of the β2 integrin subfamily are composed of an alternative α (CD11a-d) subunit, which determines the specific receptor properties, and a constant β (CD18) subunit. This review aims to present insight into the multiple immunological roles of integrin receptors, with a focus on β2 integrins that are specifically expressed by leukocytes. The pathophysiological role of β2 integrins is confirmed by the drastic phenotype of patients suffering from leukocyte adhesion deficiencies, most often resulting in severe recurrent infections and, at the same time, a predisposition for autoimmune diseases. So far, studies on the role of β2 integrins in vivo employed mice with a constitutive knockout of all β2 integrins or either family member, respectively, which complicated the differentiation between the direct and indirect effects of β2 integrin deficiency for distinct cell types. The recent generation and characterization of transgenic mice with a cell-type-specific knockdown of β2 integrins by our group has enabled the dissection of cell-specific roles of β2 integrins. Further, integrin receptors have been recognized as target receptors for the treatment of inflammatory diseases as well as tumor therapy. However, whereas both agonistic and antagonistic agents yielded beneficial effects in animal models, the success of clinical trials was limited in most cases and was associated with unwanted side effects. This unfavorable outcome is most probably related to the systemic effects of the used compounds on all leukocytes, thereby emphasizing the need to develop formulations that target distinct types of leukocytes to modulate β2 integrin activity for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (T.K.); (C.H.); (M.B.)
| |
Collapse
|
3
|
Schwab N, Wiendl H. Learning CNS immunopathology from therapeutic interventions. Sci Transl Med 2023; 15:eadg7863. [PMID: 37939164 DOI: 10.1126/scitranslmed.adg7863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/15/2023] [Indexed: 11/10/2023]
Abstract
Modulation of immune cell trafficking across the blood-brain barrier has not only introduced a therapeutic avenue for multiple sclerosis (MS) but also represents an example of reverse translational medicine. Data from clinical trials of drugs such as natalizumab and fingolimod have revealed the involvement of different compartments in relapsing versus non-relapsing MS immune biology, contributed to our understanding of central nervous system (CNS) immune surveillance, and stimulated new fields of research. Here, we discuss the results of these trials, as well as patient biomaterial-based scientific projects, and how both have informed our understanding of CNS immunopathology.
Collapse
Affiliation(s)
- Nicholas Schwab
- Department of Neurology with Institute of Translational Neurology, University of Muenster, Muenster 48149, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Muenster, Muenster 48149, Germany
- Brain and Mind Centre, University of Sydney, Camperdown NSW 2050, Australia
| |
Collapse
|
4
|
Gordon H, Rodger B, Lindsay JO, Stagg AJ. Recruitment and Residence of Intestinal T Cells - Lessons for Therapy in Inflammatory Bowel Disease. J Crohns Colitis 2023; 17:1326-1341. [PMID: 36806613 DOI: 10.1093/ecco-jcc/jjad027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Indexed: 02/23/2023]
Abstract
Targeting leukocyte trafficking in the management of inflammatory bowel disease [IBD] has been a significant therapeutic advance over the past 15 years. However, as with other advanced therapies, phase III clinical trials report response to trafficking inhibitors in only a proportion of patients, with fewer achieving clinical remission or mucosal healing. Additionally, there have been significant side effects, most notably progressive multifocal leukoencephalopathy in association with the α4 inhibitor natalizumab. This article reviews the mechanisms underpinning T cell recruitment and residence, to provide a background from which the strength and limitations of agents that disrupt leukocyte trafficking can be further explored. The therapeutic impact of trafficking inhibitors is underpinned by the complexity and plasticity of the intestinal immune response. Pathways essential for gut homing in health may be bypassed in the inflamed gut, thus providing alternative routes of entry when conventional homing molecules are targeted. Furthermore, there is conservation of trafficking architecture between proinflammatory and regulatory T cells. The persistence of resident memory cells within the gut gives rise to local established pro-inflammatory populations, uninfluenced by inhibition of trafficking. Finally, trafficking inhibitors may give rise to effects beyond the intended response, such as the impact of vedolizumab on innate immunity, as well as on target side effects. With significant research efforts into predictive biomarkers already underway, it is ultimately hoped that a better understanding of trafficking and residence will help us predict which patients are most likely to respond to inhibition of leukocyte trafficking, and how best to combine therapies.
Collapse
Affiliation(s)
- Hannah Gordon
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine, Barts & The London Medical School, Queen Mary University of London, London, UK
- Department of Gastroenterology, Barts Health NHS Trust, London, UK
| | - Beverley Rodger
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine, Barts & The London Medical School, Queen Mary University of London, London, UK
| | - James O Lindsay
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine, Barts & The London Medical School, Queen Mary University of London, London, UK
- Department of Gastroenterology, Barts Health NHS Trust, London, UK
| | - Andrew J Stagg
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine, Barts & The London Medical School, Queen Mary University of London, London, UK
| |
Collapse
|
5
|
Soldati S, Bär A, Vladymyrov M, Glavin D, McGrath JL, Gosselet F, Nishihara H, Goelz S, Engelhardt B. High levels of endothelial ICAM-1 prohibit natalizumab mediated abrogation of CD4 + T cell arrest on the inflamed BBB under flow in vitro. J Neuroinflammation 2023; 20:123. [PMID: 37221552 DOI: 10.1186/s12974-023-02797-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/02/2023] [Indexed: 05/25/2023] Open
Abstract
INTRODUCTION The humanized anti-α4 integrin blocking antibody natalizumab (NTZ) is an effective treatment for relapsing-remitting multiple sclerosis (RRMS) that is associated with the risk of progressive multifocal leukoencephalopathy (PML). While extended interval dosing (EID) of NTZ reduces the risk for PML, the minimal dose of NTZ required to maintain its therapeutic efficacy remains unknown. OBJECTIVE Here we aimed to identify the minimal NTZ concentration required to inhibit the arrest of human effector/memory CD4+ T cell subsets or of PBMCs to the blood-brain barrier (BBB) under physiological flow in vitro. RESULTS Making use of three different human in vitro BBB models and in vitro live-cell imaging we observed that NTZ mediated inhibition of α4-integrins failed to abrogate T cell arrest to the inflamed BBB under physiological flow. Complete inhibition of shear resistant T cell arrest required additional inhibition of β2-integrins, which correlated with a strong upregulation of endothelial intercellular adhesion molecule (ICAM)-1 on the respective BBB models investigated. Indeed, NTZ mediated inhibition of shear resistant T cell arrest to combinations of immobilized recombinant vascular cell adhesion molecule (VCAM)-1 and ICAM-1 was abrogated in the presence of tenfold higher molar concentrations of ICAM-1 over VCAM-1. Also, monovalent NTZ was less potent than bivalent NTZ in inhibiting T cell arrest to VCAM-1 under physiological flow. In accordance with our previous observations ICAM-1 but not VCAM-1 mediated T cell crawling against the direction of flow. CONCLUSION Taken together, our in vitro observations show that high levels of endothelial ICAM-1 abrogate NTZ mediated inhibition of T cell interaction with the BBB. EID of NTZ in MS patients may thus require consideration of the inflammatory status of the BBB as high levels of ICAM-1 may provide an alternative molecular cue allowing for pathogenic T cell entry into the CNS in the presence of NTZ.
Collapse
Affiliation(s)
- Sasha Soldati
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Alexander Bär
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Mykhailo Vladymyrov
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Dale Glavin
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory, University of Artois, Lens, France
| | - Hideaki Nishihara
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
- Department of Neurotherapeutics, Yamaguchi University, Yamaguchi, Japan
| | | | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland.
| |
Collapse
|
6
|
Konen FF, Möhn N, Witte T, Schefzyk M, Wiestler M, Lovric S, Hufendiek K, Schwenkenbecher P, Sühs KW, Friese MA, Klotz L, Pul R, Pawlitzki M, Hagin D, Kleinschnitz C, Meuth SG, Skripuletz T. Treatment of autoimmunity: The impact of disease-modifying therapies in multiple sclerosis and comorbid autoimmune disorders. Autoimmun Rev 2023; 22:103312. [PMID: 36924922 DOI: 10.1016/j.autrev.2023.103312] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
More than 10 disease-modifying therapies (DMT) are approved by the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) for the treatment of multiple sclerosis (MS) and new therapeutic options are on the horizon. Due to different underlying therapeutic mechanisms, a more individualized selection of DMTs in MS is possible, taking into account the patient's current situation. Therefore, concomitant treatment of various comorbid conditions, including autoimmune mediated disorders such as rheumatoid arthritis, should be considered in MS patients. Because the pathomechanisms of autoimmunity partially overlap, DMT could also treat concomitant inflammatory diseases and simplify the patient's treatment. In contrast, the exacerbation and even new occurrence of several autoimmune diseases have been reported as a result of immunomodulatory treatment of MS. To simplify treatment and avoid disease exacerbation, knowledge of the beneficial and adverse effects of DMT in other autoimmune disorders is critical. Therefore, we conducted a literature search and described the beneficial and adverse effects of approved and currently studied DMT in a large number of comorbid autoimmune diseases, including rheumatoid arthritis, ankylosing spondylitis, inflammatory bowel diseases, cutaneous disorders including psoriasis, Sjögren´s syndrome, systemic lupus erythematosus, systemic vasculitis, autoimmune hepatitis, and ocular autoimmune disorders. Our review aims to facilitate the selection of an appropriate DMT in patients with MS and comorbid autoimmune diseases.
Collapse
Affiliation(s)
- Franz Felix Konen
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Torsten Witte
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, 30625 Hannover, Germany..
| | - Matthias Schefzyk
- Department of Dermatology, Allergology and Venerology, Hannover Medical School, 30625 Hannover, Germany..
| | - Miriam Wiestler
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany.
| | - Svjetlana Lovric
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany.
| | - Karsten Hufendiek
- University Eye Hospital, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Kurt-Wolfram Sühs
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany.
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany.
| | - Refik Pul
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen 45147, Germany.
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, University of Tel Aviv, 6 Weizmann St., Tel-Aviv 6423906, Israel.
| | - Christoph Kleinschnitz
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen 45147, Germany.
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| |
Collapse
|
7
|
Blythe EN, Weaver LC, Brown A, Dekaban GA. β2 Integrin CD11d/CD18: From Expression to an Emerging Role in Staged Leukocyte Migration. Front Immunol 2021; 12:775447. [PMID: 34858434 PMCID: PMC8630586 DOI: 10.3389/fimmu.2021.775447] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
CD11d/CD18 is the most recently discovered and least understood β2 integrin. Known CD11d adhesive mechanisms contribute to both extravasation and mesenchymal migration – two key aspects for localizing peripheral leukocytes to sites of inflammation. Differential expression of CD11d induces differences in monocyte/macrophage mesenchymal migration including impacts on macrophage sub-set migration. The participation of CD11d/CD18 in leukocyte localization during atherosclerosis and following neurotrauma has sparked interest in the development of CD11d-targeted therapeutic agents. Whereas the adhesive properties of CD11d have undergone investigation, the signalling pathways induced by ligand binding remain largely undefined. Underlining each adhesive and signalling function, CD11d is under unique transcriptional control and expressed on a sub-set of predominately tissue-differentiated innate leukocytes. The following review is the first to capture the nearly three decades of CD11d research and discusses the emerging role of CD11d in leukocyte migration and retention during the progression of a staged immune response.
Collapse
Affiliation(s)
- Eoin N Blythe
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, ON, Canada.,Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Lynne C Weaver
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, ON, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Arthur Brown
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, ON, Canada.,Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Gregory A Dekaban
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, ON, Canada.,Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
8
|
Marques E, Paluch Z, Boháč P, Slanař O, Běláček J, Hercogová J. The safety profile of biologic agents in comparison with non-biologic systemic agents, and topical compounds in the management of psoriasis-A 30-month prospective, observational cohort study. Int J Clin Pract 2021; 75:e14915. [PMID: 34551188 DOI: 10.1111/ijcp.14915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Although biologic agents (BAs) are very effective, solid data proving they are safer than other therapies in psoriasis are still lacking. METHODS A total of 289 psoriatic patients were followed for 30 months; of which number 118 were treated with topical agents alone, 112 received BAs, and the remaining 59 patients were on non-biologic systemic agents (NBSAs). The rates of adverse events in these groups were recorded and statistically analysed. RESULTS Patients treated with BAs had higher rates of adverse events (P = .017), including overall infections (P = .003), respiratory infections (P < .001), renal, urinary (P < .001), musculoskeletal, connective tissue (P < .001, and P = .021) and oral cavity-related (P = .046) disorders. Except for the incidence of infections, all the above adverse events occurred more often in our study than in clinical trials. The occurrence of serious adverse events was P = .066, with the incidence of serious infections being P = .164. Unlike patients on topical therapy and NBSAs, patients treated with BAs were forced to discontinue their therapies (P = .001). The Psoriasis Area Severity Index (PASI) and body surface area (BSA) scores were the lowest among patients on BAs. CONCLUSION While BAs were the most effective therapies, they were associated with higher rates of treatment discontinuation and adverse events in comparison with other forms of therapy.
Collapse
Affiliation(s)
- Emanuel Marques
- Department of Dermatovenereology, Third Faculty of Medicine, Charles University and Králosvské Vinohrady University Hospital, Prague, Czech Republic
- Department of Pharmacology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Zoltán Paluch
- Department of Pharmacology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- St. John Nepomucene Neumann Institute, Příbram, Czech Republic, St. Elisabeth University of Health Care and Social Work, Bratislava, Slovak Republic
| | - Petr Boháč
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University and Bulovka University Hospital, Prague, Czech Republic
| | - Ondřej Slanař
- Department of Pharmacology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jaromír Běláček
- Department of Statistics and Operation Analysis, Faculty of Business and Economics of Brno, Mendel University in Brno, Brno, Czech Republic
| | - Jana Hercogová
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University and Bulovka University Hospital, Prague, Czech Republic
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
9
|
Bui A, Liu J, Hong J, Hadeler E, Mosca M, Brownstone N, Liao W. Identifying Novel Psoriatic Disease Drug Targets Using a Genetics-Based Priority Index Pipeline. JOURNAL OF PSORIASIS AND PSORIATIC ARTHRITIS 2021; 6:185-197. [PMID: 35756599 PMCID: PMC9229908 DOI: 10.1177/24755303211026023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
BACKGROUND Despite numerous genome-wide association studies conducted in psoriasis and psoriatic arthritis, only a small fraction of the identified genes has been therapeutically targeted. OBJECTIVE We sought to identify and analyze potential therapeutic targets for psoriasis and psoriatic arthritis (PsA) using the priority index (Pi), a genetics-dependent drug target prioritization approach. METHODS Significant genetic variants from GWAS for psoriasis, PsA, and combined psoriatic disease were annotated and run through the Pi pipeline. Potential drug targets were identified based on genomic predictors, annotation predictors, pathway enrichment, and pathway crosstalk. RESULTS Several gene targets were identified for psoriasis and PsA that demonstrated biological associations to their respective diseases. Some are currently being explored as potential therapeutic targets (i.e. ICAM1, NF-kB, REV3L, ADRA1B for psoriasis; CCL11 for PsA); others have not yet been investigated (i.e. LNPEP, LCE3 for psoriasis; UBLCP1 for PsA). Additionally, many nodal points of potential intervention were identified as promising therapeutic targets. Of these, some are currently being studied such as TYK2 for psoriasis, and others have yet to be explored (i.e. PPP2CA, YAP1, PI3K, AKT, FOXO1, RELA, CSF2, IFNGR1, IFNGR2 for psoriasis; GNAQ, PLCB1, GNAI2 for PsA). CONCLUSION Through Pi, we identified data-driven candidate therapeutic gene targets and pathways for psoriasis and PsA. Given the sparse PsA specific genetic studies and PsA specific drug targets, this analysis could prove to be particularly valuable in the pipeline for novel psoriatic therapies.
Collapse
Affiliation(s)
- Audrey Bui
- Department of Dermatology, University of California, San Francisco, CA 94015
- Department of Biology, St. Bonaventure University, St. Bonaventure, NY 14778
| | - Jared Liu
- Department of Dermatology, University of California, San Francisco, CA 94015
| | - Julie Hong
- Department of Dermatology, University of California, San Francisco, CA 94015
| | - Edward Hadeler
- Department of Dermatology, University of California, San Francisco, CA 94015
| | - Megan Mosca
- Department of Dermatology, University of California, San Francisco, CA 94015
| | - Nicholas Brownstone
- Department of Dermatology, University of California, San Francisco, CA 94015
| | - Wilson Liao
- Department of Dermatology, University of California, San Francisco, CA 94015
| |
Collapse
|
10
|
Frenken P, Hartung HP, Olsson T, Adams O, Warnke C. Type O blood group associates with higher anti-JC polyomavirus antibody levels. Brain Behav 2021; 11:e2298. [PMID: 34291599 PMCID: PMC8413794 DOI: 10.1002/brb3.2298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/27/2021] [Accepted: 07/09/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Patients with multiple sclerosis (MS) and high anti-JC polyomavirus (JCPyV) antibodies in blood have an increased risk for the development of progressive multifocal leukoencephalopathy (PML) when treated for MS. To test the hypothesis that type O blood group associates with anti-JCPyV antibody levels and the risk of developing PML, we characterized ABO blood group antigen on blood samples of 62 patients with PML, and 64 MS controls without PML. METHODS Monocentric retrospective cohort study. Anti-JCPyV antibody levels in arbitrary units (AU) were determined in sera using an ELISA-based method, and blood group specific antibodies using standardised test erythrocytes. RESULTS Anti-JCPyV antibody levels were higher in individuals with blood group O compared with all other blood groups (O: median AU: 129; not O: median AU: 53; p = .005). This association was not observed for the closely related BK virus. Of the 62 patients with PML, 29 (47%, 95% confidence interval (CI) 35%-59%) were of blood group O, which showed a nonsignificant trend to differ from the expected distribution in the German population (41%), and the MS controls studied (36%, 95% CI 25%-48%). CONCLUSION The ABO blood group O antigen associates with higher anti-JCPyV antibody levels and may impact the risk of the later development of PML. The overrepresentation of blood group O in cases with PML was in line with a previous publication. Larger studies are warranted to assess a potential value of host genetic markers, such as the ABO status, for PML risk prediction during immunotherapy.
Collapse
Affiliation(s)
- Pia Frenken
- Institute for Virologie, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tomas Olsson
- Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
| | - Ortwin Adams
- Institute for Virologie, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Clemens Warnke
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.,Department of Neurology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
11
|
Koritzinsky EH, Tsuda H, Fairchild RL. Endogenous memory T cells with donor-reactivity: early post-transplant mediators of acute graft injury in unsensitized recipients. Transpl Int 2021; 34:1360-1373. [PMID: 33963616 PMCID: PMC8389524 DOI: 10.1111/tri.13900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/15/2021] [Accepted: 05/03/2021] [Indexed: 11/29/2022]
Abstract
The pretransplant presence of endogenous donor-reactive memory T cells is an established risk factor for acute rejection and poorer transplant outcomes. A major source of these memory T cells in unsensitized recipients is heterologously generated memory T cells expressing reactivity to donor allogeneic MHC molecules. Multiple clinical studies have shown that the pretransplant presence of high numbers of circulating endogenous donor-reactive memory T cells correlates with higher incidence of acute rejection and decreased graft function during the first-year post-transplant. These findings have spurred investigation in preclinical models to better understand mechanisms underlying endogenous donor-reactive memory T-cell-mediated allograft injury in unsensitized graft recipients. These studies have led to the identification of unique mechanisms underlying the activation of these memory T cells within allografts at early times after transplant. In particular, optimal activation to mediate acute allograft injury is dependent on the intensity of ischaemia-reperfusion injury. Therapeutic strategies directed at the recruitment and activation of endogenous donor-reactive memory T cells are effective in attenuating acute injury in allografts experiencing increased ischaemia-reperfusion injury in preclinical models and should be translatable to clinical transplantation.
Collapse
Affiliation(s)
- Erik H. Koritzinsky
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Hidetoshi Tsuda
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Robert L. Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Transplant Center, Cleveland Clinic, Cleveland, OH
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| |
Collapse
|
12
|
A Case of John Cunningham Virus Induced Rhombencephalitis after Rituximab Therapy for Idiopathic Thrombocytopenic Purpura. Case Rep Infect Dis 2021; 2021:5525053. [PMID: 34221521 PMCID: PMC8219458 DOI: 10.1155/2021/5525053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 06/05/2021] [Indexed: 11/17/2022] Open
Abstract
Background John Cunningham virus (JCV) is known to cause progressive multifocal leukoencephalopathy (PML) in immuno-compromised patients due to lytic infection of oligodendrocytes and astrocytes. Rarely, it may also present as granule cell neuronopathy (GCN), leading to degeneration of cerebellar granule cell neurons. It is described in patients with underlying conditions or medication contributing to immune compromise. Case Presentation. A 73-year-old man presented with ataxia and difficulty in speech which began 3 months after initiation of treatment for idiopathic thrombocytopenic purpura with rituximab. Neurological examination was significant for torsional nystagmus, motor aphasia, right-sided dysmetria, and dysdiadochokinesia with gait ataxia. Magnetic resonance imaging (MRI) showed right cerebellar lesion and cerebrospinal fluid (CSF) polymerase chain reaction (PCR) was positive for JC virus. Conclusion The diagnosis of JC virus-related cerebellar disease can be missed, due to the subacute to chronic onset and challenges in detection. Clinicians should have a high degree of suspicion for development of these symptoms, even a few months after initiation of immune-modulatory therapy because the progression and outcomes can be disastrous.
Collapse
|
13
|
Rosenbaum JT, Harrington CA, Searles RP, Fei SS, Zaki A, Arepalli S, Paley MA, Hassman LM, Vitale AT, Conrady CD, Keath P, Mitchell C, Watson L, Planck SR, Martin TM, Choi D. Identifying RNA Biomarkers and Molecular Pathways Involved in Multiple Subtypes of Uveitis. Am J Ophthalmol 2021; 226:226-234. [PMID: 33503442 DOI: 10.1016/j.ajo.2021.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Uveitis is a heterogeneous collection of diseases. We tested the hypothesis that despite the diversity of uveitides, there could be common mechanisms shared by multiple subtypes, and that evidence of these common mechanisms may be detected as gene expression profiles in whole blood. DESIGN Cohort study. METHODS Ninety subjects with uveitis including axial spondyloarthritis (n = 17), sarcoidosis (n = 13), inflammatory bowel disease (n = 12), tubulointerstitial nephritis with uveitis (n = 10), or idiopathic uveitis (n = 38) as well as 18 healthy controls were enrolled, predominantly at Oregon Health & Science University. RNA-Seq data generated from peripheral, whole blood identified 19,859 unique transcripts. We analyzed gene expression pathways via Kyoto Encyclopedia of Genes and Genomes and Gene Ontology (GO). We validated our list of upregulated genes by comparison to a previously published study on peripheral blood gene expression among 50 subjects with diverse forms of uveitis. RESULTS Both the Kyoto Encyclopedia of Genes and Genomes and GO analysis identified multiple shared pathways or GO terms with a P value of <.0001. Almost all pathways related to the immune response and/or response to an infection. A total of 119 individual transcripts were upregulated by at least 1.5-fold and false discovery rate <.05, and 61 were downregulated by similar criteria. Comparing mRNA from our study with a false discovery rate <.05 and the prior report, we identified 10 common gene transcripts: ICAM1, IL15RA, IL15, IRF1, IL10RB, GSK3A, TYK2, MEF2A, MEF2B, and MEF2D. CONCLUSIONS Many forms of uveitis share overlapping mechanisms. These data support the concept that a single therapeutic approach could benefit diverse forms of this disease.
Collapse
|
14
|
Abstract
The risk of JC polyomavirus encephalopathy varies among biologic classes and among agents within the same class. Of currently used biologics, the highest risk is seen with natalizumab followed by rituximab. Multiple other agents have also been implicated. Drug-specific causality is difficult to establish because many patients receive multiple immunomodulatory medications concomitantly or sequentially, and have other immunocompromising factors related to their underlying disease. As use of biologic therapies continues to expand, further research is needed into pathogenesis, treatment, and prevention of JC polyomavirus encephalopathy such that risk for its development is better understood and mitigated, if not eliminated altogether.
Collapse
|
15
|
Shao S, Wang G, Maverakis E, Gudjonsson JE. Targeted Treatment for Erythrodermic Psoriasis: Rationale and Recent Advances. Drugs 2020; 80:525-534. [PMID: 32180204 DOI: 10.1007/s40265-020-01283-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Erythrodermic psoriasis (EP) is an extreme and often refractory variant of psoriasis with high morbidity and increased mortality, and is frequently classified as a dermatological emergency. The pathophysiology of EP is largely unknown but is thought to differ from that of plaque psoriasis. Treatment of EP is challenging, and usually based on clinical experience and patient co-morbidities, due to its low incidence and limited clinical evidence. Conventional treatments, such as topical glucocorticoid therapy, cyclosporin, acitretin, and methotrexate have some but limited efficacy in EP, and treatment discontinuation may result in flares. Newer biological drugs, including anti-TNF, anti-IL-17, and anti-IL-12/23 agents, have shown promise in therapeutic management of EP, but most of the available evidence is currently based on small case series and reports. Few studies have compared available treatment options for EP, and further clinical studies are necessary to provide clinical data and optimal treatment guidelines for EP patients. Here, we provide a comprehensive review of the background of EP, assess the available clinical data on the efficacy of targeted therapies, and aim to provide a foundation for clinical decision making for this rare form of psoriasis.
Collapse
Affiliation(s)
- Shuai Shao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, 710032, China
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, 710032, China
| | - Emanual Maverakis
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, 95616, USA
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
16
|
Perro M, Iannacone M, von Andrian UH, Peixoto A. Role of LFA-1 integrin in the control of a lymphocytic choriomeningitis virus (LCMV) infection. Virulence 2020; 11:1640-1655. [PMID: 33251934 PMCID: PMC7714442 DOI: 10.1080/21505594.2020.1845506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Leukocyte function-associated antigen 1 (LFA-1) is the most widely expressed member of the β2 integrin family of cell-cell adhesion molecules. Although LFA-1 is thought to regulate multiple aspects of T cell immunity, its role in the response of CD8+ T cells to viral infections remains unclear. Indeed, compelling clinical evidence shows that loss of LFA-1 function predisposes to infection in humans but animal models show limited to no susceptibility to infection. Here, we addressed this conundrum in a mouse model of infection with lymphocytic choriomeningitis virus (LCMV), where CD8+ T cells are necessary and sufficient to confer protection. To this end, we followed the fate and function of wild-type and LFA-1 deficient virus-specific CD8+ T cells and assessed the effect of blocking anti-LFA-1 monoclonal antibody in the outcome of infection. Our analysis of viral clearance and T cell responses using transcriptome profiling reveals a role for LFA-1 as a gatekeeper of effector T cell survival and dysfunction that when defective can predispose to LCMV infection.
Collapse
Affiliation(s)
- Mario Perro
- Harvard Medical School , Department of Microbiology and Immunobiology, Boston, Massachusetts, USA
| | - Matteo Iannacone
- Harvard Medical School , Department of Microbiology and Immunobiology, Boston, Massachusetts, USA
| | - Ulrich H von Andrian
- Harvard Medical School , Department of Microbiology and Immunobiology, Boston, Massachusetts, USA
| | - Antonio Peixoto
- Harvard Medical School , Department of Microbiology and Immunobiology, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Lauver MD, Lukacher AE. JCPyV VP1 Mutations in Progressive MultifocalLeukoencephalopathy: Altering Tropismor Mediating Immune Evasion? Viruses 2020; 12:v12101156. [PMID: 33053912 PMCID: PMC7600905 DOI: 10.3390/v12101156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022] Open
Abstract
Polyomaviruses are ubiquitous human pathogens that cause lifelong, asymptomatic infections in healthy individuals. Although these viruses are restrained by an intact immune system, immunocompromised individuals are at risk for developing severe diseases driven by resurgent viral replication. In particular, loss of immune control over JC polyomavirus can lead to the development of the demyelinating brain disease progressive multifocal leukoencephalopathy (PML). Viral isolates from PML patients frequently carry point mutations in the major capsid protein, VP1, which mediates virion binding to cellular glycan receptors. Because polyomaviruses are non-enveloped, VP1 is also the target of the host's neutralizing antibody response. Thus, VP1 mutations could affect tropism and/or recognition by polyomavirus-specific antibodies. How these mutations predispose susceptible individuals to PML and other JCPyV-associated CNS diseases remains to be fully elucidated. Here, we review the current understanding of polyomavirus capsid mutations and their effects on viral tropism, immune evasion, and virulence.
Collapse
|
18
|
Lauver MD, Goetschius DJ, Netherby-Winslow CS, Ayers KN, Jin G, Haas DG, Frost EL, Cho SH, Bator CM, Bywaters SM, Christensen ND, Hafenstein SL, Lukacher AE. Antibody escape by polyomavirus capsid mutation facilitates neurovirulence. eLife 2020; 9:e61056. [PMID: 32940605 PMCID: PMC7541085 DOI: 10.7554/elife.61056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/17/2020] [Indexed: 12/27/2022] Open
Abstract
JCPyV polyomavirus, a member of the human virome, causes progressive multifocal leukoencephalopathy (PML), an oft-fatal demyelinating brain disease in individuals receiving immunomodulatory therapies. Mutations in the major viral capsid protein, VP1, are common in JCPyV from PML patients (JCPyV-PML) but whether they confer neurovirulence or escape from virus-neutralizing antibody (nAb) in vivo is unknown. A mouse polyomavirus (MuPyV) with a sequence-equivalent JCPyV-PML VP1 mutation replicated poorly in the kidney, a major reservoir for JCPyV persistence, but retained the CNS infectivity, cell tropism, and neuropathology of the parental virus. This mutation rendered MuPyV resistant to a monoclonal Ab (mAb), whose specificity overlapped the endogenous anti-VP1 response. Using cryo-EM and a custom sub-particle refinement approach, we resolved an MuPyV:Fab complex map to 3.2 Å resolution. The structure revealed the mechanism of mAb evasion. Our findings demonstrate convergence between nAb evasion and CNS neurovirulence in vivo by a frequent JCPyV-PML VP1 mutation.
Collapse
Affiliation(s)
- Matthew D Lauver
- Department of Microbiology and Immunology, Penn State College of MedicineHersheyUnited States
| | - Daniel J Goetschius
- Department of Biochemistry and Molecular Biology, Pennsylvania State UniversityUniversity ParkUnited States
| | | | - Katelyn N Ayers
- Department of Microbiology and Immunology, Penn State College of MedicineHersheyUnited States
| | - Ge Jin
- Department of Microbiology and Immunology, Penn State College of MedicineHersheyUnited States
| | - Daniel G Haas
- Department of Microbiology and Immunology, Penn State College of MedicineHersheyUnited States
| | - Elizabeth L Frost
- Department of Microbiology and Immunology, Penn State College of MedicineHersheyUnited States
| | - Sung Hyun Cho
- Huck Institutes of the Life Sciences, Pennsylvania State UniversityUniversity ParkUnited States
| | - Carol M Bator
- Huck Institutes of the Life Sciences, Pennsylvania State UniversityUniversity ParkUnited States
| | - Stephanie M Bywaters
- Department of Pathology, Penn State College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
| | - Neil D Christensen
- Department of Pathology, Penn State College of MedicineHersheyUnited States
- The Jake Gittlen Laboratories for Cancer Research, Penn State College of MedicineHersheyUnited States
| | - Susan L Hafenstein
- Department of Biochemistry and Molecular Biology, Pennsylvania State UniversityUniversity ParkUnited States
- Huck Institutes of the Life Sciences, Pennsylvania State UniversityUniversity ParkUnited States
- Department of Medicine, Penn State College of MedicineHersheyUnited States
| | - Aron E Lukacher
- Department of Microbiology and Immunology, Penn State College of MedicineHersheyUnited States
| |
Collapse
|
19
|
Abstract
INTRODUCTION Integrins are a family of 24 cell adhesion receptors that play a role in the biggest unmet needs in medicine - cardiovascular disease, immunology and cancer. Their discovery promised huge potential for the pharmaceutical industry. Areas covered. Over 35-years since their discovery, there is little to show for the hundreds of billions of dollars of investment in anti-integrin drug discovery programmes. In this review the author discusses the reasons for the failure of this promising class of drugs and the future for this class of drugs. Expert opinion. Within 10-years, there was a plethora of potent, specific anti-integrin molecules and since their discovery, many of these agents have entered clinical trials. The success in discovering these agents was due to recently discovered monoclonal antibody technology. The integrin-recognition domain Arg-Gly-Asp (RGD) provided the basis for discovering small molecule inhibitors to integrins - both cyclic peptides and peptidomimetics. Most agents failed in the Phase III clinical trials and those agents that did make it to the market were plagued with issues of toxicity and limited efficacy and were soon replaced with non-integrin targeting agents. Their failure was due to a combination of poor pharmacokinetics and pharmacodynamics, complicated by the complex pathophysiology of integrins.
Collapse
Affiliation(s)
- Dermot Cox
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland , Dublin, Ireland
| |
Collapse
|
20
|
Abstract
Lymphocyte depletion and blockade of T-cell activation and trafficking serve as therapeutic strategies for an enlarging number of immune-mediated diseases and malignancies. This review summarizes the infection risks associated to monoclonal antibodies that bind to the α chain of the interleukin-2 receptor, the cell surface glycoprotein CD52, and members of α4- and β2-integrin families acting as cell-adhesion molecules. An outline of the mechanisms of action, approved indications and off-label uses, expected impact on the host immune response, and available clinical evidence is provided for each of these agents.
Collapse
|
21
|
Carballido JM, Regairaz C, Rauld C, Raad L, Picard D, Kammüller M. The Emerging Jamboree of Transformative Therapies for Autoimmune Diseases. Front Immunol 2020; 11:472. [PMID: 32296421 PMCID: PMC7137386 DOI: 10.3389/fimmu.2020.00472] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
Standard treatments for autoimmune and autoinflammatory disorders rely mainly on immunosuppression. These are predominantly symptomatic remedies that do not affect the root cause of the disease and are associated with multiple side effects. Immunotherapies are being developed during the last decades as more specific and safer alternatives to small molecules with broad immunosuppressive activity, but they still do not distinguish between disease-causing and protective cell targets and thus, they still have considerable risks of increasing susceptibility to infections and/or malignancy. Antigen-specific approaches inducing immune tolerance represent an emerging trend carrying the potential to be curative without inducing broad immunosuppression. These therapies are based on antigenic epitopes derived from the same proteins that are targeted by the autoreactive T and B cells, and which are administered to patients together with precise instructions to induce regulatory responses capable to restore homeostasis. They are not personalized medicines, and they do not need to be. They are precision therapies exquisitely targeting the disease-causing cells that drive pathology in defined patient populations. Immune tolerance approaches are truly transformative options for people suffering from autoimmune diseases.
Collapse
Affiliation(s)
- José M. Carballido
- Translational Medicine, Novartis Institutes for Biomedical Research, Basel, Switzerland
- Autoimmunity Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Camille Regairaz
- Autoimmunity Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Celine Rauld
- Autoimmunity Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Layla Raad
- Autoimmunity Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Damien Picard
- Translational Medicine, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Michael Kammüller
- Translational Medicine, Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
22
|
Bednarczyk M, Stege H, Grabbe S, Bros M. β2 Integrins-Multi-Functional Leukocyte Receptors in Health and Disease. Int J Mol Sci 2020; 21:E1402. [PMID: 32092981 PMCID: PMC7073085 DOI: 10.3390/ijms21041402] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 12/25/2022] Open
Abstract
β2 integrins are heterodimeric surface receptors composed of a variable α (CD11a-CD11d) and a constant β (CD18) subunit and are specifically expressed by leukocytes. The α subunit defines the individual functional properties of the corresponding β2 integrin, but all β2 integrins show functional overlap. They mediate adhesion to other cells and to components of the extracellular matrix (ECM), orchestrate uptake of extracellular material like complement-opsonized pathogens, control cytoskeletal organization, and modulate cell signaling. This review aims to delineate the tremendous role of β2 integrins for immune functions as exemplified by the phenotype of LAD-I (leukocyte adhesion deficiency 1) patients that suffer from strong recurrent infections. These immune defects have been largely attributed to impaired migratory and phagocytic properties of polymorphonuclear granulocytes. The molecular base for this inherited disease is a functional impairment of β2 integrins due to mutations within the CD18 gene. LAD-I patients are also predisposed for autoimmune diseases. In agreement, polymorphisms within the CD11b gene have been associated with autoimmunity. Consequently, β2 integrins have received growing interest as targets in the treatment of autoimmune diseases. Moreover, β2 integrin activity on leukocytes has been implicated in tumor development.
Collapse
Affiliation(s)
| | | | | | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (H.S.); (S.G.)
| |
Collapse
|
23
|
Nishihara H, Soldati S, Mossu A, Rosito M, Rudolph H, Muller WA, Latorre D, Sallusto F, Sospedra M, Martin R, Ishikawa H, Tenenbaum T, Schroten H, Gosselet F, Engelhardt B. Human CD4 + T cell subsets differ in their abilities to cross endothelial and epithelial brain barriers in vitro. Fluids Barriers CNS 2020; 17:3. [PMID: 32008573 PMCID: PMC6996191 DOI: 10.1186/s12987-019-0165-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Background The brain barriers establish compartments in the central nervous system (CNS) that significantly differ in their communication with the peripheral immune system. In this function they strictly control T-cell entry into the CNS. T cells can reach the CNS by either crossing the endothelial blood–brain barrier (BBB) or the epithelial blood-cerebrospinal fluid barrier (BCSFB) of the choroid plexus (ChP). Objective Analysis of the cellular and molecular mechanisms involved in the migration of different human CD4+ T-cell subsets across the BBB versus the BCSFB. Methods Human in vitro models of the BBB and BCSFB were employed to study the migration of circulating and CNS-entry experienced CD4+ T helper cell subsets (Th1, Th1*, Th2, Th17) across the BBB and BCSFB under inflammatory and non-inflammatory conditions in vitro. Results While under non-inflammatory conditions Th1* and Th1 cells preferentially crossed the BBB, under inflammatory conditions the migration rate of all Th subsets across the BBB was comparable. The migration of all Th subsets across the BCSFB from the same donor was 10- to 20-fold lower when compared to their migration across the BBB. Interestingly, Th17 cells preferentially crossed the BCSFB under both, non-inflamed and inflamed conditions. Barrier-crossing experienced Th cells sorted from CSF of MS patients showed migratory characteristics indistinguishable from those of circulating Th cells of healthy donors. All Th cell subsets could additionally cross the BCSFB from the CSF to ChP stroma side. T-cell migration across the BCSFB involved epithelial ICAM-1 irrespective of the direction of migration. Conclusions Our observations underscore that different Th subsets may use different anatomical routes to enter the CNS during immune surveillance versus neuroinflammation with the BCSFB establishing a tighter barrier for T-cell entry into the CNS compared to the BBB. In addition, CNS-entry experienced Th cell subsets isolated from the CSF of MS patients do not show an increased ability to cross the brain barriers when compared to circulating Th cell subsets from healthy donors underscoring the active role of the brain barriers in controlling T-cell entry into the CNS. Also we identify ICAM-1 to mediate T cell migration across the BCSFB.
Collapse
Affiliation(s)
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Adrien Mossu
- Theodor Kocher Institute, University of Bern, Bern, Switzerland.,Transcure Bioservices, Archamps, France
| | - Maria Rosito
- Theodor Kocher Institute, University of Bern, Bern, Switzerland.,Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Henriette Rudolph
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - William A Muller
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Daniela Latorre
- Institute for Research in Biomedicine, Università Della Svizzera Italiana, Bellinzona, Switzerland.,Institute for Microbiology, ETH Zurich, Zurich, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università Della Svizzera Italiana, Bellinzona, Switzerland.,Institute for Microbiology, ETH Zurich, Zurich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tobias Tenenbaum
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Horst Schroten
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Fabien Gosselet
- Blood Brain Barrier Laboratory, University of Artois, Lens, France
| | | |
Collapse
|
24
|
Khalili A, Craigie M, Donadoni M, Sariyer IK. Host-Immune Interactions in JC Virus Reactivation and Development of Progressive Multifocal Leukoencephalopathy (PML). J Neuroimmune Pharmacol 2019; 14:649-660. [PMID: 31452013 PMCID: PMC6898772 DOI: 10.1007/s11481-019-09877-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022]
Abstract
With the advent of immunomodulatory therapies and the HIV epidemic, the impact of JC Virus (JCV) on the public health system has grown significantly due to the increased incidence of Progressive Multifocal Leukoencephalopathy (PML). Currently, there are no pharmaceutical agents targeting JCV infection for the treatment and the prevention of viral reactivation leading to the development of PML. As JCV primarily reactivates in immunocompromised patients, it is proposed that the immune system (mainly the cellular-immunity component) plays a key role in the regulation of JCV to prevent productive infection and PML development. However, the exact mechanism of JCV immune regulation and reactivation is not well understood. Likewise, the impact of host factors on JCV regulation and reactivation is another understudied area. Here we discuss the current literature on host factor-mediated and immune factor-mediated regulation of JCV gene expression with the purpose of developing a model of the factors that are bypassed during JCV reactivation, and thus are potential targets for the development of therapeutic interventions to suppress PML initiation. Graphical Abstract.
Collapse
Affiliation(s)
- Amir Khalili
- Department of Neuroscience and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 North Broad Street, Medical Education and Research Building, 7th Floor, Philadelphia, PA, 19140, USA
| | - Michael Craigie
- Department of Neuroscience and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 North Broad Street, Medical Education and Research Building, 7th Floor, Philadelphia, PA, 19140, USA
| | - Martina Donadoni
- Department of Neuroscience and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 North Broad Street, Medical Education and Research Building, 7th Floor, Philadelphia, PA, 19140, USA
| | - Ilker Kudret Sariyer
- Department of Neuroscience and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 North Broad Street, Medical Education and Research Building, 7th Floor, Philadelphia, PA, 19140, USA.
| |
Collapse
|
25
|
Verma NK, Chalasani MLS, Scott JD, Kelleher D. CG-NAP/Kinase Interactions Fine-Tune T Cell Functions. Front Immunol 2019; 10:2642. [PMID: 31781123 PMCID: PMC6861388 DOI: 10.3389/fimmu.2019.02642] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/24/2019] [Indexed: 01/04/2023] Open
Abstract
CG-NAP, also known as AKAP450, is an anchoring/adaptor protein that streamlines signal transduction in various cell types by localizing signaling proteins and enzymes with their substrates. Great efforts are being devoted to elucidating functional roles of this protein and associated macromolecular signaling complex. Increasing understanding of pathways involved in regulating T lymphocytes suggests that CG-NAP can facilitate dynamic interactions between kinases and their substrates and thus fine-tune T cell motility and effector functions. As a result, new binding partners of CG-NAP are continually being uncovered. Here, we review recent advances in CG-NAP research, focusing on its interactions with kinases in T cells with an emphasis on the possible role of this anchoring protein as a target for therapeutic intervention in immune-mediated diseases.
Collapse
Affiliation(s)
- Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | | | - John D Scott
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, United States
| | - Dermot Kelleher
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore.,Departments of Medicine and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
26
|
Sinnecker T, Hadisurya J, Schneider-Hohendorf T, Schwab N, Wrede K, Gembruch O, Gold R, Hellwig K, Pilgram-Pastor S, Adams O, Albrecht P, Hartung HP, Aktas O, Kraemer M. Extensive immune reconstitution inflammatory syndrome in Fingolimod-associated PML: a case report with 7 Tesla MRI data. BMC Neurol 2019; 19:190. [PMID: 31399069 PMCID: PMC6688281 DOI: 10.1186/s12883-019-1407-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 07/19/2019] [Indexed: 11/10/2022] Open
Abstract
Background Progressive multifocal leukoencephalopathy (PML) is a rare complication of patients treated with fingolimod. Case presentation Routine MRI eventually led to diagnosis of asymptomatic early PML that remained stable after discontinuation of fingolimod. As blood lymphocyte counts normalized, signs of immune reconstitution inflammatory syndrome (IRIS) and renewed MS activity developed. Both, advanced laboratory and ultrahigh field MRI findings elucidated differences between PML and MS. Conclusions In our case, early discontinuation of fingolimod yielded a good outcome, lymphocyte counts reflected immune system activity, and paraclinical findings helped to differentiate between PML-IRIS and MS.
Collapse
Affiliation(s)
- Tim Sinnecker
- Department of Neurology, Universitätsspital, Basel, Switzerland.,Medical Image Analysis Center Basel, Basel, Switzerland
| | - Jeffrie Hadisurya
- Department of Neurology, Alfried Krupp von Bohlen und Halbach Hospital, Alfried-Krupp-Str. 21, 45117, Essen, Germany
| | - Tilman Schneider-Hohendorf
- Clinic of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Nicholas Schwab
- Clinic of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Karsten Wrede
- Department of Neurosurgery, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Erwin L. Hahn Institute for Magnetic Resonance Imaging, University Duisburg-Essen, Essen, Germany
| | - Oliver Gembruch
- Department of Neurosurgery, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Erwin L. Hahn Institute for Magnetic Resonance Imaging, University Duisburg-Essen, Essen, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Kerstin Hellwig
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | | | - Ortwin Adams
- Institute of Virology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Markus Kraemer
- Department of Neurology, Alfried Krupp von Bohlen und Halbach Hospital, Alfried-Krupp-Str. 21, 45117, Essen, Germany. .,Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
27
|
Bakaeva T, Prasad S. For Massachusetts Eye and Ear Special Issue: Updates on Therapies for Multiple Sclerosis for the Ophthalmologist. Semin Ophthalmol 2019; 34:270-278. [PMID: 31158038 DOI: 10.1080/08820538.2019.1620806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In the past decade, the available disease-modifying therapies for multiple sclerosis have broadened significantly, providing physicians and patients with multiple options with different mechanisms of action, administration routes, and risk-benefit profiles. Multiple sclerosis often presents with ophthalmic manifestations due to inflammatory demyelination of the afferent and efferent visual pathways, and evidence of disease can factor into the decision to initiate or substitute a particular therapy. Furthermore, some of these drugs have toxicities that can manifest with ophthalmic complications, of which ophthalmologists should be aware.
Collapse
Affiliation(s)
- Tatiana Bakaeva
- a Massachusetts Eye and Ear Infirmary , Harvard Medical School , Boston , MA , USA
| | - Sashank Prasad
- b Brigham and Women's Hospital , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
28
|
Haghi Navand A, Teimoori A, Makvandi M, Nisi N, Seyedian SS, Ranjbari N, Ahmadi Angali K, Keyani H, Tabasi M, Pourjabari K. Study on JV Virus in Patients with Colon Cancer Type
Adenocarcinoma. Asian Pac J Cancer Prev 2019; 20:1147-1151. [PMID: 31030488 PMCID: PMC6948910 DOI: 10.31557/apjcp.2019.20.4.1147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Introduction: Colorectal cancer is the most repetitious malignancies with high mortality worldwide. JC virus (JCV) is ubiquitous Polyomavirus, with seroprevalence rates ranging from 70% to 90% in adult population. Recently the role of JCV have been reported in many malignant tumors worldwide. The association of JCV was reported in patients with colon and rectum cancers. Thus this study was conducted to evaluate the association of JCV DNA in patients with colon cancer type Adenocarcinoma. Material and Methods: A total of 120 formalin-fixed paraffin-embedded tissue blocks samples were collected including 20/40(50%) males, 20/40(50%) females patients with Colorectal Cancer(CRC), and 80 (50% males, 50% females) patients with benign tumor as a control. DNA was extracted for all the samples. Nested PCR was carried out for detection of Vp1/T-Ag junction genome in JCV genome by Nested-PCR assay. Randomly, PCR products of 6 samples were sequenced to analysis the partial JCV DNA. The phylogeny tree was constructed to determine homology identity with other JCV. Results: 4/40(10%) samples of test group and 10/80 (12.5%) of control samples were positive for JCV DNA (P= 0.69). Out of 4 samples positive for JC DNA, 3(7.5%) were males and 1(2.4%) female (P=0.29). The frequency of JCV DNA in age group> 50 years was 4/32(10%), while in age group <50 years was 0/8 (0%) (p= 0.29). Conclusion: prevalence of JCV DNA was among 10% patients with CRC and 12.5% benign tumors (p=0.69). The distribution of JCV DNA was among 7.5% male and 2.5% female (p= 0.29). The frequency of JCV DNA was among 10% cases of age group >50 years and 0% of age group <50 years (P= 0.29). The subsequent T-Ag protein expression might explain the increased risk of colorectal cancer and requires further investigation.
Collapse
Affiliation(s)
- Azadeh Haghi Navand
- Infectious and Tropical Diseases Research Center Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. manoochehrmakvandi299@ gmail.com,Virology Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Teimoori
- Virology Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Manoochehr Makvandi
- Infectious and Tropical Diseases Research Center Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. manoochehrmakvandi299@ gmail.com,Virology Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nilofar Nisi
- Infectious and Tropical Diseases Research Center Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. manoochehrmakvandi299@ gmail.com,Virology Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Saeid Seyedian
- Alimentary Tract Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nastarn Ranjbari
- Department of Pathology, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kambiz Ahmadi Angali
- Department of Biostatistic, School of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadis Keyani
- Infectious and Tropical Diseases Research Center Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. manoochehrmakvandi299@ gmail.com
| | - Maryam Tabasi
- Infectious and Tropical Diseases Research Center Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. manoochehrmakvandi299@ gmail.com
| | - Keyvan Pourjabari
- Infectious and Tropical Diseases Research Center Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. manoochehrmakvandi299@ gmail.com
| |
Collapse
|
29
|
Blankenbach K, Schwab N, Hofner B, Adams O, Keller-Stanislawski B, Warnke C. Natalizumab-associated progressive multifocal leukoencephalopathy in Germany. Neurology 2019; 92:e2232-e2239. [PMID: 30952796 DOI: 10.1212/wnl.0000000000007451] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 01/09/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To evaluate characteristics relevant to diagnosis of JC polyomavirus-associated progressive multifocal leukoencephalopathy (PML), and PML risk stratification in a large national cohort of patients with multiple sclerosis during therapy with natalizumab. METHODS Analysis of 292 adverse drug reaction forms on suspected cases of PML reported to the German national competent authority until July 2017. Patients not fulfilling PML diagnostic criteria or with insufficient information available were excluded. RESULTS Of the 142 confirmed patients with PML, 72.3% (95% confidence interval [CI] 64.4%-79.1%) were women, and the median age was 43 years (range 19-69). Of these patients, 7.7% (95% CI 4.3%-13.5%) were clinically asymptomatic at time of PML diagnosis. PML was fatal in 9.1% (95% CI 5.3%-15.1%) of the patients. Infratentorial lesions on imaging were reported in 40% (95% CI 32.0%-48.6%) of the patients. JC polyomavirus DNA in CSF was undetectable at time of first analysis in 23.8% (95% CI 17.3%-31.9%) of the patients. Three patients tested negative for anti-JC polyomavirus antibodies within 6 to 18 months before PML diagnosis, with seroconversion confirmed 5.5 months, 7 months (in a post hoc analysis only), or at time of PML diagnosis. CONCLUSIONS JC polyomavirus DNA detection in CSF has limited sensitivity in early PML, and clinical and imaging presentation may be atypical. Thus, critical revision of current PML diagnostic criteria is warranted. Negative anti-JC polyomavirus antibodies in sera do not preclude the later development of PML. This emphasizes the need for close and regular serologic, imaging, and clinical monitoring in patients treated with natalizumab.
Collapse
Affiliation(s)
- Kira Blankenbach
- From the Department Safety of Medicinal Products and Medical Devices (K.B., B.K.-S.), and Section Biostatistics (B.H.), Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen; Clinic of Neurology with Institute of Translational Neurology (N.S.), University Hospital Muenster, University Muenster; Institute for Virology (O.A.), University of Duesseldorf, Medical Faculty, Duesseldorf; and Department of Neurology (C.W.), University Hospital Koeln, Germany
| | - Nicholas Schwab
- From the Department Safety of Medicinal Products and Medical Devices (K.B., B.K.-S.), and Section Biostatistics (B.H.), Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen; Clinic of Neurology with Institute of Translational Neurology (N.S.), University Hospital Muenster, University Muenster; Institute for Virology (O.A.), University of Duesseldorf, Medical Faculty, Duesseldorf; and Department of Neurology (C.W.), University Hospital Koeln, Germany
| | - Benjamin Hofner
- From the Department Safety of Medicinal Products and Medical Devices (K.B., B.K.-S.), and Section Biostatistics (B.H.), Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen; Clinic of Neurology with Institute of Translational Neurology (N.S.), University Hospital Muenster, University Muenster; Institute for Virology (O.A.), University of Duesseldorf, Medical Faculty, Duesseldorf; and Department of Neurology (C.W.), University Hospital Koeln, Germany
| | - Ortwin Adams
- From the Department Safety of Medicinal Products and Medical Devices (K.B., B.K.-S.), and Section Biostatistics (B.H.), Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen; Clinic of Neurology with Institute of Translational Neurology (N.S.), University Hospital Muenster, University Muenster; Institute for Virology (O.A.), University of Duesseldorf, Medical Faculty, Duesseldorf; and Department of Neurology (C.W.), University Hospital Koeln, Germany
| | - Brigitte Keller-Stanislawski
- From the Department Safety of Medicinal Products and Medical Devices (K.B., B.K.-S.), and Section Biostatistics (B.H.), Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen; Clinic of Neurology with Institute of Translational Neurology (N.S.), University Hospital Muenster, University Muenster; Institute for Virology (O.A.), University of Duesseldorf, Medical Faculty, Duesseldorf; and Department of Neurology (C.W.), University Hospital Koeln, Germany
| | - Clemens Warnke
- From the Department Safety of Medicinal Products and Medical Devices (K.B., B.K.-S.), and Section Biostatistics (B.H.), Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen; Clinic of Neurology with Institute of Translational Neurology (N.S.), University Hospital Muenster, University Muenster; Institute for Virology (O.A.), University of Duesseldorf, Medical Faculty, Duesseldorf; and Department of Neurology (C.W.), University Hospital Koeln, Germany.
| |
Collapse
|
30
|
|
31
|
Parasramani S, Pillai J. Biologics in psoriasis: Indian experience. INDIAN JOURNAL OF DRUGS IN DERMATOLOGY 2019. [DOI: 10.4103/ijdd.ijdd_33_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
32
|
Sârbu MI, Georgescu SR, Tampa M, Sârbu AE, Simionescu O. Biological therapies in psoriasis - revisited. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2018; 56:75-84. [PMID: 29168976 DOI: 10.1515/rjim-2017-0045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Indexed: 06/07/2023]
Abstract
Psoriasis is a chronic, immune mediated disorder affecting approximately 2% of the population. Even in our days, patients with psoriasis are confronted with stigmatization and social rejection. As a result, their quality of life is significantly impaired. Biological therapies have revolutionized the treatment of moderate to severe psoriasis. The aim of this paper is to look over the most important biological therapies available for the management of plaque-type psoriasis.
Collapse
Affiliation(s)
| | - Simona-Roxana Georgescu
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- "Victor Babeş" Hospital of Infectious and Tropical Diseases, Dermatology Department, Bucharest, Romania
| | - Mircea Tampa
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- "Victor Babeş" Hospital of Infectious and Tropical Diseases, Dermatology Department, Bucharest, Romania
| | | | - Olga Simionescu
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- "Colentina" Clinical Hospital, Dermatology Department I, Bucharest, Romania
| |
Collapse
|
33
|
Walling BL, Kim M. LFA-1 in T Cell Migration and Differentiation. Front Immunol 2018; 9:952. [PMID: 29774029 PMCID: PMC5943560 DOI: 10.3389/fimmu.2018.00952] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/17/2018] [Indexed: 01/21/2023] Open
Abstract
Maintenance of homeostatic immune surveillance and development of effective adaptive immune responses require precise regulation of spatial and temporal lymphocyte trafficking throughout the body to ensure pathogen clearance and memory generation. Dysregulation of lymphocyte activation and migration can lead to impaired adaptive immunity, recurrent infections, and an array of autoimmune diseases and chronic inflammation. Central to the recruitment of T cells, integrins are cell surface receptors that regulate adhesion, signal transduction, and migration. With 24 integrin pairs having been discovered to date, integrins are defined not only by the composition of the heterodimeric pair but by cell-type specific expression and their ligands. Furthermore, integrins not only facilitate adhesion but also induce intracellular signaling and have recently been uncovered as mechanosensors providing additional complexity to the signaling pathways. Among several leukocyte-specific integrins, lymphocyte function-associated antigen-1 (LFA-1 or αLβ2; CD11a/CD18) is a key T cell integrin, which plays a major role in regulating T cell activation and migration. Adhesion to LFA-1's ligand, intracellular adhesion receptor 1 (ICAM-1) facilitates firm endothelium adhesion, prolonged contact with antigen-presenting cells, and binding to target cells for killing. While the downstream signaling pathways utilized by LFA-1 are vastly conserved they allow for highly disparate responses. Here, we summarize the roles of LFA-1 and ongoing studies to better understand its functions and regulation.
Collapse
Affiliation(s)
- Brandon L Walling
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
34
|
Maillart E, Taoufik Y, Gasnault J, Stankoff B. Leucoencefalopatia multifocale progressiva. Neurologia 2018. [DOI: 10.1016/s1634-7072(18)89404-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
35
|
Redelman-Sidi G, Michielin O, Cervera C, Ribi C, Aguado JM, Fernández-Ruiz M, Manuel O. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Immune checkpoint inhibitors, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors). Clin Microbiol Infect 2018; 24 Suppl 2:S95-S107. [PMID: 29427804 DOI: 10.1016/j.cmi.2018.01.030] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/18/2018] [Accepted: 01/27/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND The present review is part of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Infections in Compromised Hosts (ESGICH) consensus document on the safety of targeted and biological therapies. AIMS To review, from an infectious diseases perspective, the safety profile of immune checkpoint inhibitors, LFA-3-targeted agents, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors, and to suggest preventive recommendations. SOURCES Computer-based Medline searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death (PD)-1/PD-1 ligand 1 (PD-L1)-targeted agents do not appear to intrinsically increase the risk of infection but can induce immune-related adverse effects requiring additional immunosuppression. Although CD4+ T-cell lymphopenia is associated with alefacept, no opportunistic infections have been observed. Progressive multifocal leukoencephalopathy (PML) may occur during therapy with natalizumab (anti-α4-integrin monoclonal antibody (mAb)) and efalizumab (anti-CD11a mAb), but no cases have been reported to date with vedolizumab (anti-α4β7 mAb). In patients at high risk for PML (positive anti-JC polyomavirus serology with serum antibody index >1.5 and duration of therapy ≥48 months), the benefit-risk ratio of continuing natalizumab should be carefully considered. Fingolimod induces profound peripheral blood lymphopenia and increases the risk of varicella zoster virus (VZV) infection. Prophylaxis with (val)acyclovir and VZV vaccination should be considered. Proteasome inhibitors also increase the risk of VZV infection, and antiviral prophylaxis with (val)acyclovir is recommended. Anti-Pneumocystis prophylaxis may be considered in myeloma multiple patients with additional risk factors (i.e. high-dose corticosteroids). IMPLICATIONS Clinicians should be aware of the risk of immune-related adverse effects and PML in patients receiving immune checkpoint and cell adhesion inhibitors respectively.
Collapse
Affiliation(s)
- G Redelman-Sidi
- Service of Infectious Disease, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA.
| | - O Michielin
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - C Cervera
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - C Ribi
- Department of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - J M Aguado
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i+12), Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - M Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i+12), Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - O Manuel
- Department of Infectious Diseases, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
36
|
Schittenhelm L, Hilkens CM, Morrison VL. β 2 Integrins As Regulators of Dendritic Cell, Monocyte, and Macrophage Function. Front Immunol 2017; 8:1866. [PMID: 29326724 PMCID: PMC5742326 DOI: 10.3389/fimmu.2017.01866] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/08/2017] [Indexed: 12/24/2022] Open
Abstract
Emerging evidence suggests that the β2 integrin family of adhesion molecules have an important role in suppressing immune activation and inflammation. β2 integrins are important adhesion and signaling molecules that are exclusively expressed on leukocytes. The four β2 integrins (CD11a, CD11b, CD11c, and CD11d paired with the β2 chain CD18) play important roles in regulating three key aspects of immune cell function: recruitment to sites of inflammation; cell-cell contact formation; and downstream effects on cellular signaling. Through these three processes, β2 integrins both contribute to and regulate immune responses. This review explores the pro- and anti-inflammatory effects of β2 integrins in monocytes, macrophages, and dendritic cells and how they influence the outcome of immune responses. We furthermore discuss how imbalances in β2 integrin function can have far-reaching effects on mounting appropriate immune responses, potentially influencing the development and progression of autoimmune and inflammatory diseases. Therapeutic targeting of β2 integrins, therefore, holds enormous potential in exploring treatment options for a variety of inflammatory conditions.
Collapse
Affiliation(s)
- Leonie Schittenhelm
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom.,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, United Kingdom
| | - Catharien M Hilkens
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, United Kingdom
| | - Vicky L Morrison
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, United Kingdom
| |
Collapse
|
37
|
Breuer J, Herich S, Schneider-Hohendorf T, Chasan AI, Wettschureck N, Gross CC, Loser K, Zarbock A, Roth J, Klotz L, Wiendl H, Schwab N. Dual action by fumaric acid esters synergistically reduces adhesion to human endothelium. Mult Scler 2017; 24:1871-1882. [PMID: 28984166 DOI: 10.1177/1352458517735189] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Dimethyl fumarate (DMF) is prescribed against relapsing-remitting multiple sclerosis (MS). Here, we investigated the effects of DMF and monomethyl fumarate (MMF), its metabolite in vivo, at the (inflamed) blood-brain barrier (BBB). METHODS Effects of fumaric acid esters were analyzed using primary human brain-derived microvascular endothelial cells (HBMECs) in combination with peripheral blood mononuclear cells (PBMCs) derived from DMF-treated MS patients. RESULTS MMF-binding to brain endothelium cells leads to activation of nuclear factor (erythroid-derived 2)-related factor 2 (Nrf2)-induced downregulation of vascular cell adhesion molecule 1 (VCAM-1). This might be mediated via the G-protein-coupled receptor (GPCR) hydroxycarboxylic acid receptor 2 (HCA2), a known molecular target of MMF, as we could demonstrate its expression and regulation on HBMECs. DMF treatment in vivo led to a strongly reduced expression of VCAM-1's ligand very late antigen 4 (VLA-4) by selectively reducing integrin high-expressing memory T cells of MS patients, potentially due to inhibition of their maturation by reduced trans-localization of NFκB. CONCLUSION DMF-mediated VCAM-1 downregulation on the endothelial side and reduction in T cells with a migratory phenotype on the lymphocyte side result in a synergistic reduction in T-cell adhesion to activated endothelium and, therefore, to reduced BBB transmigration in the setting of MS.
Collapse
Affiliation(s)
- Johanna Breuer
- Department of Neurology, University of Münster, Münster, Germany
| | - Sebastian Herich
- Department of Neurology, University of Münster, Münster, Germany
| | | | - Achmet I Chasan
- Institute of Immunology, University of Münster, Münster, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany/Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Karin Loser
- Department of Dermatology, University of Münster, Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Luisa Klotz
- Department of Neurology, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | - Nicholas Schwab
- Department of Neurology, University of Münster, Münster, Germany
| |
Collapse
|
38
|
PML risk and natalizumab: the elephant in the room. Lancet Neurol 2017; 16:864-865. [PMID: 28969985 DOI: 10.1016/s1474-4422(17)30335-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/19/2017] [Indexed: 01/21/2023]
|
39
|
Veenstra M, Williams DW, Calderon TM, Anastos K, Morgello S, Berman JW. Frontline Science: CXCR7 mediates CD14 +CD16 + monocyte transmigration across the blood brain barrier: a potential therapeutic target for NeuroAIDS. J Leukoc Biol 2017; 102:1173-1185. [PMID: 28754798 DOI: 10.1189/jlb.3hi0517-167r] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/22/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
CD14+CD16+ monocytes transmigrate into the CNS of HIV-positive people in response to chemokines elevated in the brains of infected individuals, including CXCL12. Entry of these cells leads to viral reservoirs, neuroinflammation, and neuronal damage. These may eventually lead to HIV-associated neurocognitive disorders. Although antiretroviral therapy (ART) has significantly improved the lives of HIV-infected people, the prevalence of cognitive deficits remains unchanged despite ART, still affecting >50% of infected individuals. There are no therapies to reduce these deficits or to prevent CNS entry of CD14+CD16+ monocytes. The goal of this study was to determine whether CXCR7, a receptor for CXCL12, is expressed on CD14+CD16+ monocytes and whether a small molecule CXCR7 antagonist (CCX771) can prevent CD14+CD16+ monocyte transmigration into the CNS. We showed for the first time that CXCR7 is on CD14+CD16+ monocytes and that it may be a therapeutic target to reduce their entry into the brain. We demonstrated that CD14+CD16+ monocytes and not the more abundant CD14+CD16- monocytes or T cells transmigrate to low homeostatic levels of CXCL12. This may be a result of increased CXCR7 on CD14+CD16+ monocytes. We showed that CCX771 reduced transmigration of CD14+CD16+ monocytes but not of CD14+CD16- monocytes from uninfected and HIV-infected individuals and that it reduced CXCL12-mediated chemotaxis of CD14+CD16+ monocytes. We propose that CXCR7 is a therapeutic target on CD14+CD16+ monocytes to limit their CNS entry, thereby reducing neuroinflammation, neuronal damage, and HIV-associated neurocognitive disorders. Our data also suggest that CCX771 may reduce CD14+CD16+ monocyte-mediated inflammation in other disorders.
Collapse
Affiliation(s)
- Mike Veenstra
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Dionna W Williams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tina M Calderon
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kathryn Anastos
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Susan Morgello
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; and
| | - Joan W Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA; .,Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
40
|
Misbah SA. Progressive multi-focal leucoencephalopathy - driven from rarity to clinical mainstream by iatrogenic immunodeficiency. Clin Exp Immunol 2017; 188:342-352. [PMID: 28245526 PMCID: PMC5422720 DOI: 10.1111/cei.12948] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2017] [Indexed: 12/21/2022] Open
Abstract
Advances in immune-mediated targeted therapies have proved to be a double-edged sword for patients by highlighting the risk of iatrogenic infective complications. This has been exemplified by progressive multi-focal leucoencephalopathy (PML), a hitherto rare devastating viral infection of the brain caused by the neurotrophic JC polyoma virus. While PML achieved prominence during the first two decades of the HIV epidemic, effective anti-retroviral treatment and restitution of T cell function has led to PML being less prominent in this population. HIV infection as a predisposing factor has now been supplanted by T cell immunodeficiency induced by a range of immune-mediated therapies as a major cause of PML. This review focuses on PML in the context of therapeutic immunosuppression and encompasses therapeutic monoclonal antibodies, novel immunomodulatory agents such as Fingolimod and dimethyl fumarate, as well as emerging data on PML in primary immune deficiency.
Collapse
Affiliation(s)
- S A Misbah
- Department of Clinical Immunology, Oxford University Hospitals, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
41
|
Henrickson SE, Ruffner MA, Kwan M. Unintended Immunological Consequences of Biologic Therapy. Curr Allergy Asthma Rep 2017; 16:46. [PMID: 27324478 DOI: 10.1007/s11882-016-0624-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recent advances in the understanding of immune dysregulation in autoimmune diseases have enabled the development of new monoclonal antibody-based drugs called biologics. Biologics have been used to target aberrant immune responses in many diseases, but patients with rheumatologic and other autoimmune diseases have benefited the most and improvements in outcomes have been significant. The use of biologics is not without hazard, however, as these agents block immune pathways adapted to protect the host. This has been borne out by increased rates of infections as well as induction of new autoimmune and hematologic adverse effects. As new drugs for the treatment of autoimmune conditions are entering the pipeline, it is incumbent on the practicing immunologist to understand the mechanism of these biologics and the implications of clinical use.
Collapse
Affiliation(s)
- Sarah E Henrickson
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, 3550 Market St. 3rd floor, Philadelphia, PA, 19104, USA
| | - Melanie A Ruffner
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, 3550 Market St. 3rd floor, Philadelphia, PA, 19104, USA
| | - Mildred Kwan
- Department of Internal Medicine, Division of Rheumatology, Allergy & Immunology, University of North Carolina School of Medicine, 3300 Thurston, CB #7280, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
42
|
Davis BP, Ballas ZK. Biologic response modifiers: Indications, implications, and insights. J Allergy Clin Immunol 2017; 139:1445-1456. [PMID: 28263774 DOI: 10.1016/j.jaci.2017.02.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/03/2017] [Accepted: 02/09/2017] [Indexed: 12/27/2022]
Abstract
The field of biologic immune modulators is currently mushrooming at a dizzying pace. Although most of these biologics are tested and approved for one or a few indications, their unanticipated side effects and off-label use have contributed significantly to our understanding of basic immune mechanisms, the involvement of cytokines in several apparently nonimmunologic diseases, and the importance of compartmentalized immune responses. In this review we attempt to give a bird's-eye view of the major biologics and to highlight insights and implications derived from their secondary effects and adverse reactions.
Collapse
Affiliation(s)
- Benjamin P Davis
- Department of Internal Medicine, Division of Immunology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.
| | - Zuhair K Ballas
- Department of Internal Medicine, Division of Immunology, University of Iowa and the Iowa City Veterans Administration, Iowa City, Iowa
| |
Collapse
|
43
|
Nowacki M, Nazarewski Ł, Kloskowski T, Tyloch D, Pokrywczyńska M, Pietkun K, Jundziłł A, Tyloch J, Habib SL, Drewa T. Novel surgical techniques, regenerative medicine, tissue engineering and innovative immunosuppression in kidney transplantation. Arch Med Sci 2016; 12:1158-1173. [PMID: 27695507 PMCID: PMC5016594 DOI: 10.5114/aoms.2016.61919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 02/08/2015] [Indexed: 01/09/2023] Open
Abstract
On the 60th anniversary of the first successfully performed renal transplantation, we summarize the historical, current and potential future status of kidney transplantation. We discuss three different aspects with a potential significant influence on kidney transplantation progress: the development of surgical techniques, the influence of regenerative medicine and tissue engineering, and changes in immunosuppression. We evaluate the standard open surgical procedures with modern techniques and compare them to less invasive videoscopic as well as robotic techniques. The role of tissue engineering and regenerative medicine as a potential method for future kidney regeneration or replacement and the interesting search for novel solutions in the field of immunosuppression will be discussed. After 60 years since the first successfully performed kidney transplantation, we can conclude that the greatest achievements are associated with the development of surgical techniques and with planned systemic immunosuppression.
Collapse
Affiliation(s)
- Maciej Nowacki
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
- Chair of Surgical Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Łukasz Nazarewski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Kloskowski
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Dominik Tyloch
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Marta Pokrywczyńska
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Katarzyna Pietkun
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Arkadiusz Jundziłł
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Janusz Tyloch
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Samy L. Habib
- Department of Geriatrics, Geriatric Research, Education, and Clinical Center, South Texas Veterans Healthcare System, San Antonio, TX, USA
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Tomasz Drewa
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
- Department of General and Oncological Urology, Nicolaus Copernicus Hospital, Torun, Poland
| |
Collapse
|
44
|
Leukocyte adhesion molecule dynamics after Natalizumab withdrawal in Multiple Sclerosis. Clin Immunol 2016; 171:18-24. [DOI: 10.1016/j.clim.2016.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 11/20/2022]
|
45
|
Wharton KA, Quigley C, Themeles M, Dunstan RW, Doyle K, Cahir-McFarland E, Wei J, Buko A, Reid CE, Sun C, Carmillo P, Sur G, Carulli JP, Mansfield KG, Westmoreland SV, Staugaitis SM, Fox RJ, Meier W, Goelz SE. JC Polyomavirus Abundance and Distribution in Progressive Multifocal Leukoencephalopathy (PML) Brain Tissue Implicates Myelin Sheath in Intracerebral Dissemination of Infection. PLoS One 2016; 11:e0155897. [PMID: 27191595 PMCID: PMC4871437 DOI: 10.1371/journal.pone.0155897] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 05/05/2016] [Indexed: 12/22/2022] Open
Abstract
Over half of adults are seropositive for JC polyomavirus (JCV), but rare individuals develop progressive multifocal leukoencephalopathy (PML), a demyelinating JCV infection of the central nervous system. Previously, PML was primarily seen in immunosuppressed patients with AIDS or certain cancers, but it has recently emerged as a drug safety issue through its association with diverse immunomodulatory therapies. To better understand the relationship between the JCV life cycle and PML pathology, we studied autopsy brain tissue from a 70-year-old psoriasis patient on the integrin alpha-L inhibitor efalizumab following a ~2 month clinical course of PML. Sequence analysis of lesional brain tissue identified PML-associated viral mutations in regulatory (non-coding control region) DNA, capsid protein VP1, and the regulatory agnoprotein, as well as 9 novel mutations in capsid protein VP2, indicating rampant viral evolution. Nine samples, including three gross PML lesions and normal-appearing adjacent tissues, were characterized by histopathology and subject to quantitative genomic, proteomic, and molecular localization analyses. We observed a striking correlation between the spatial extent of demyelination, axonal destruction, and dispersion of JCV along white matter myelin sheath. Our observations in this case, as well as in a case of PML-like disease in an immunocompromised rhesus macaque, suggest that long-range spread of polyomavirus and axonal destruction in PML might involve extracellular association between virus and the white matter myelin sheath.
Collapse
Affiliation(s)
- Keith A. Wharton
- Translational Pathology Laboratory, Biogen Inc., Cambridge, MA, United States of America
- * E-mail:
| | - Catherine Quigley
- Translational Pathology Laboratory, Biogen Inc., Cambridge, MA, United States of America
| | - Marian Themeles
- Translational Pathology Laboratory, Biogen Inc., Cambridge, MA, United States of America
| | - Robert W. Dunstan
- Translational Pathology Laboratory, Biogen Inc., Cambridge, MA, United States of America
| | - Kathryn Doyle
- Immunology, Biogen Inc., Cambridge, MA, United States of America
| | | | - Jing Wei
- Bioanalytical Chemistry, Biogen Inc., Cambridge, MA, United States of America
| | - Alex Buko
- Bioanalytical Chemistry, Biogen Inc., Cambridge, MA, United States of America
| | - Carl E. Reid
- Molecular Discovery, Biogen Inc., Cambridge, MA, United States of America
| | - Chao Sun
- Molecular Discovery, Biogen Inc., Cambridge, MA, United States of America
| | - Paul Carmillo
- Molecular Discovery, Biogen Inc., Cambridge, MA, United States of America
| | - Gargi Sur
- Molecular Discovery, Biogen Inc., Cambridge, MA, United States of America
| | - John P. Carulli
- Molecular Discovery, Biogen Inc., Cambridge, MA, United States of America
| | - Keith G. Mansfield
- Department of Pathology, Harvard Medical School, New England Primate Research Center, Southborough, MA, United States of America
| | - Susan V. Westmoreland
- Department of Pathology, Harvard Medical School, New England Primate Research Center, Southborough, MA, United States of America
| | - Susan M. Staugaitis
- Departments of Pathology, Neurosciences, and Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH, United States of America
| | - Robert J. Fox
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH, United States of America
| | - Werner Meier
- Discovery Sciences, Biogen Inc, Cambridge, MA, United States of America
| | - Susan E. Goelz
- Neurology, Biogen Inc, Cambridge, MA, United States of America
| |
Collapse
|
46
|
Abstract
Antigen-experienced T cells, also known as memory T cells, are functionally and phenotypically distinct from naive T cells. Their enhanced expression of adhesion molecules and reduced requirement for co-stimulation enables them to mount potent and rapid recall responses to subsequent antigen encounters. Memory T cells generated in response to prior antigen exposures can cross-react with other nonidentical, but similar, antigens. This heterologous cross-reactivity not only enhances protective immune responses, but also engenders de novo alloimmunity. This latter characteristic is increasingly recognized as a potential barrier to allograft acceptance that is worthy of immunotherapeutic intervention, and several approaches have been investigated. Calcineurin inhibition effectively controls memory T-cell responses to allografts, but this benefit comes at the expense of increased infectious morbidity. Lymphocyte depletion eliminates allospecific T cells but spares memory T cells to some extent, such that patients do not completely lose protective immunity. Co-stimulation blockade is associated with reduced adverse-effect profiles and improved graft function relative to calcineurin inhibition, but lacks efficacy in controlling memory T-cell responses. Targeting the adhesion molecules that are upregulated on memory T cells might offer additional means to control co-stimulation-blockade-resistant memory T-cell responses.
Collapse
|
47
|
Kampylafka EI, Alexopoulos H, Dalakas MC, Tzioufas AG. Immunotherapies for Neurological Manifestations in the Context of Systemic Autoimmunity. Neurotherapeutics 2016; 13:163-78. [PMID: 26510559 PMCID: PMC4720664 DOI: 10.1007/s13311-015-0393-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Neurological involvement is relatively common in the majority of systemic autoimmune diseases and may lead to severe morbidity and mortality, if not promptly treated. Treatment options vary greatly, depending on the underlying systemic pathophysiology and the associated neurological symptoms. Selecting the appropriate therapeutic scheme is further complicated by the lack of definite therapeutic guidelines, the necessity to differentiate primary neurological syndromes from those related to the underlying systemic disease, and to sort out adverse neurological manifestations caused by immunosuppressants or the biological agents used to treat the primary disease. Immunotherapy is a sine qua non for treating most, if not all, neurological conditions presenting in the context of systemic autoimmunity. Specific agents include classical immune modulators such as corticosteroids, cyclophosphamide, intravenous immunoglobulin, and plasma exchange, as well as numerous biological therapies, for example anti-tumor necrosis factor agents and monoclonal antibodies that target various immune pathways such as B cells, cytokines, and co-stimulatory molecules. However, experience regarding the use of these agents in neurological complications of systemic diseases is mainly empirical or based on small uncontrolled studies and case series. The aim of this review is to present the state-of-the-art therapies applied in various neurological manifestations encountered in the context of systemic autoimmune diseases; evaluate all treatment options on the basis of existing guidelines; and compliment these data with our personal experience derived from a large number of patients.
Collapse
Affiliation(s)
- Eleni I Kampylafka
- Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Harry Alexopoulos
- Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Marinos C Dalakas
- Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Athanasios G Tzioufas
- Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
| |
Collapse
|
48
|
Bittner S, Wiendl H. Neuroimmunotherapies Targeting T Cells: From Pathophysiology to Therapeutic Applications. Neurotherapeutics 2016; 13:4-19. [PMID: 26563391 PMCID: PMC4720668 DOI: 10.1007/s13311-015-0405-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Therapeutic options for multiple sclerosis (MS) have significantly increased over the last few years. T lymphocytes are considered to play a central role in initiating and perpetuating the pathological immune response. Currently approved therapies for MS target T lymphocytes, either in an unspecific manner or directly by interference with specific T-cell pathways. While the concept of "T-cell-specific therapy" implies specificity and selectivity, currently approved approaches come from a general shaping of the immune system towards anti-inflammatory immune responses by non-T-cell-selective immune suppression or immune modulation (e.g., interferons-immune modulation approach) to a depletion of immune cell populations involving T cells (e.g., anti-CD52, alemtuzumab-immune selective depletion approach), or a selective inhibition of distinct molecular pathways in order to sequester leucocytes (e.g., natalizumab-leukocyte sequestration approach). This review will highlight the rationale and results of different T-cell-directed therapeutic approaches coming from basic animal experiments to clinical trials. We will first discuss the pathophysiological rationale for targeting T lymphocytes in MS leading to currently approved treatments acting on T lymphocytes. Furthermore, we will disuss previous promising concepts that have failed to show efficacy in clinical trials or were halted as a result of unexpected adverse events. Learning from the discrepancies between expectations and failures in practical outcomes helps to optimize future research approaches and clinical study designs. As our current view of MS pathogenesis and patient needs is rapidly evolving, novel therapeutic approaches targeting T lymphocytes will also be discussed, including specific molecular interventions such as cytokine-directed treatments or strategies enhancing immunoregulatory mechanisms. Based on clinical experience and novel pathophysiological approaches, T-cell-based strategies will remain a pillarstone of MS therapy.
Collapse
Affiliation(s)
- Stefan Bittner
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Neurology, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany.
| |
Collapse
|
49
|
Kreuger J, Phillipson M. Targeting vascular and leukocyte communication in angiogenesis, inflammation and fibrosis. Nat Rev Drug Discov 2015; 15:125-42. [PMID: 26612664 DOI: 10.1038/nrd.2015.2] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulation of vascular permeability, recruitment of leukocytes from blood to tissue and angiogenesis are all processes that occur at the level of the microvasculature during both physiological and pathological conditions. The interplay between microvascular cells and leukocytes during inflammation, together with the emerging roles of leukocytes in the modulation of the angiogenic process, make leukocyte-vascular interactions prime targets for therapeutics to potentially treat a wide range of diseases, including pathological and dysfunctional vessel growth, chronic inflammation and fibrosis. In this Review, we discuss how the different cell types that are present in and around microvessels interact, cooperate and instruct each other, and in this context we highlight drug targets as well as emerging druggable processes that can be exploited to restore tissue homeostasis.
Collapse
Affiliation(s)
- Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Uppsala, 75123, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Uppsala, 75123, Sweden
| |
Collapse
|
50
|
Wollebo HS, Bellizzi A, Kaminski R, Hu W, White MK, Khalili K. CRISPR/Cas9 System as an Agent for Eliminating Polyomavirus JC Infection. PLoS One 2015; 10:e0136046. [PMID: 26360417 PMCID: PMC4567079 DOI: 10.1371/journal.pone.0136046] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/30/2015] [Indexed: 12/21/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a fatal demyelinating disease of the central nervous system (CNS) caused by reactivation of the human polyomavirus JCV gene expression and its replication in oligodendrocytes, the myelin producing cells in the brain. Once a rare disease seen in patients with lymphotproliferative and myeloproliferative disorders, PML has been seen more frequently in HIV-1 positive/AIDS patients as well as patients undergoing immunomodulatory therapy due for autoimmune disorders including multiple sclerosis, rheumatoid arthritis, and others. As of now there is no cure for PML and in most cases disease progression leads to death within two years. Similar to other polyomaviruses, the JCV genome is small circular double stranded DNA that includes coding sequences for the viral early protein, T-antigen, which is critical for directing viral reactivation and lytic infection. Here, we employ a newly developed gene editing strategy, CRISPR/Cas9, to introduce mutations in the viral genome and, by inactivating the gene encoding T-antigen, inhibit viral replication. We first used bioinformatics screening and identified several potential targets within the JCV T-antigen gene that can serve as sites for the creation of guide RNAs (gRNAs) for positioning the Cas9 nuclease on the designated area of the viral genome for editing. Results from a series of integrated genetic and functional studies showed that transient or conditional expression of Cas9 and gRNAs specifically targets the DNA sequences corresponding to the N-terminal region of T-antigen, and by introducing mutation, interferes with expression and function of of the viral protein, hence suppressing viral replication in permissive cells. Results from SURVEYOR assay revealed no off-target effects of the JCV-specific CRISPR/Cas9 editing apparatus. These observations provide the first evidence for the employment of a gene editing strategy as a promising tool for the elimination of the JCV genome and a potential cure for PML.
Collapse
MESH Headings
- Antigens, Viral, Tumor/genetics
- Base Sequence
- CRISPR-Cas Systems
- Cell Line, Tumor
- Gene Expression
- Gene Knockdown Techniques
- Gene Targeting
- Genetic Therapy/methods
- Genome, Viral
- Humans
- JC Virus/genetics
- Leukoencephalopathy, Progressive Multifocal/therapy
- Leukoencephalopathy, Progressive Multifocal/virology
- Molecular Sequence Data
- Mutation
- Promoter Regions, Genetic
- RNA Editing
- RNA, Guide, CRISPR-Cas Systems/chemistry
- RNA, Guide, CRISPR-Cas Systems/genetics
- Sequence Alignment
- Virus Replication
Collapse
Affiliation(s)
- Hassen S. Wollebo
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Anna Bellizzi
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Rafal Kaminski
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Wenhui Hu
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Martyn K. White
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Kamel Khalili
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| |
Collapse
|