1
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [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: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Marastoni D, Turano E, Tamanti A, Colato E, Pisani AI, Scartezzini A, Carotenuto S, Mazziotti V, Camera V, Anni D, Ziccardi S, Guandalini M, Pizzini FB, Virla F, Mariotti R, Magliozzi R, Bonetti B, Steinman L, Calabrese M. Association of Levels of CSF Osteopontin With Cortical Atrophy and Disability in Early Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200265. [PMID: 38917380 PMCID: PMC11203401 DOI: 10.1212/nxi.0000000000200265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/29/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND AND OBJECTIVES To evaluate CSF inflammatory markers with accumulation of cortical damage as well as disease activity in patients with early relapsing-remitting MS (RRMS). METHODS CSF levels of osteopontin (OPN) and 66 inflammatory markers were assessed using an immune-assay multiplex technique in 107 patients with RRMS (82 F/25 M, mean age 35.7 ± 11.8 years). All patients underwent regular clinical assessment and yearly 3T MRI scans for 2 years while 39 patients had a 4-year follow-up. White matter lesion number and volume, cortical lesions (CLs) and volume, and global cortical thickness (CTh) were evaluated together with the 'no evidence of disease activity' (NEDA-3) status, defined by no relapses, no disability worsening, and no MRI activity, including CLs. RESULTS The random forest algorithm selected OPN, CXCL13, TWEAK, TNF, IL19, sCD30, sTNFR1, IL35, IL16, and sCD163 as significantly associated with changes in global CTh. OPN and CXCL13 were most related to accumulation of atrophy after 2 and 4 years. In a multivariate linear regression model on CSF markers, OPN (p < 0.001), CXCL13 (p = 0.001), and sTNFR1 (p = 0.024) were increased in those patients with accumulating atrophy (adjusted R-squared 0.615). The 10 markers were added in a model that included all clinical, demographic, and MRI variables: OPN (p = 0.002) and IL19 (p = 0.022) levels were confirmed to be significantly increased in patients developing more CTh change over the follow-up (adjusted R-squared 0.619). CXCL13 and OPN also revealed the best association with NEDA-3 after 2 years, with OPN significantly linked to disability accumulation (OR 2.468 [1.46-5.034], p = 0.004) at the multivariate logistic regression model. DISCUSSION These data confirm and expand our knowledge on the prognostic role of the CSF inflammatory profile in predicting changes in cortical pathology and disease activity in early MS. The data emphasize a crucial role of OPN.
Collapse
Affiliation(s)
- Damiano Marastoni
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Ermanna Turano
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Agnese Tamanti
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Elisa Colato
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Anna Isabella Pisani
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Arianna Scartezzini
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Silvia Carotenuto
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Valentina Mazziotti
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Valentina Camera
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Daniela Anni
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Stefano Ziccardi
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Maddalena Guandalini
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Francesca B Pizzini
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Federica Virla
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Raffaella Mariotti
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Roberta Magliozzi
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Bruno Bonetti
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Lawrence Steinman
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| | - Massimiliano Calabrese
- From the Neurology B (D.M., E.T., A.T., E.C., A.I.P., A.S., S.C., V.M., V.C., D.A., S.Z., M.G., F.V., R. Magliozzi, M.C.); Anatomy and Histology section (E.T., F.V., R. Mariotti), Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy; Department of Anatomy and Neurosciences (E.C.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands; Neuroradiology and Radiology Units (F.B.P.), Department of Engineering for Innovation Medicine, University of Verona, Italy; Department of Brain Sciences (R. Magliozzi), Faculty of Medicine, Imperial College London, United Kingdom; Neurology A (B.B.), Azienda Ospedaliera Universitaria Integrata di Verona, Italy; and Department of Neurology and Neurological Sciences Stanford University (L.S.), CA
| |
Collapse
|
3
|
Goldberg Y, Segal S, Hamdi L, Nabat H, Fainstein N, Mediouni E, Asis Y, Theotokis P, Salamotas I, Grigoriadis N, Katz A, Ben-Hur T, Einstein O. High-intensity interval training attenuates development of autoimmune encephalomyelitis solely by systemic immunomodulation. Sci Rep 2023; 13:16513. [PMID: 37783693 PMCID: PMC10545672 DOI: 10.1038/s41598-023-43534-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
The impact of high-intensity interval training (HIIT) on the central nervous system (CNS) in autoimmune neuroinflammation is not known. The aim of this study was to determine the direct effects of HIIT on the CNS and development of experimental autoimmune encephalomyelitis (EAE). Healthy mice were subjected to HIIT by treadmill running and the proteolipid protein (PLP) transfer EAE model was utilized. To examine neuroprotection, PLP-reactive lymph-node cells (LNCs) were transferred to HIIT and sedentary (SED) mice. To examine immunomodulation, PLP-reactive LNCs from HIIT and SED donor mice were transferred to naïve recipients and analyzed in vitro. HIIT in recipient mice did not affect the development of EAE following exposure to PLP-reactive LNCs. HIIT mice exhibited enhanced migration of systemic autoimmune cells into the CNS and increased demyelination. In contrast, EAE severity in recipient mice injected with PLP-reactive LNCs from HIIT donor mice was significantly diminished. The latter positive effect was associated with decreased migration of autoimmune cells into the CNS and inhibition of very late antigen (VLA)-4 expression in LNCs. Thus, the beneficial effect of HIIT on EAE development is attributed solely to systemic immunomodulatory effects, likely because of systemic inhibition of autoreactive cell migration and reduced VLA-4 integrin expression.
Collapse
Affiliation(s)
- Yehuda Goldberg
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700, Ariel, Israel
| | - Shir Segal
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700, Ariel, Israel
| | - Liel Hamdi
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700, Ariel, Israel
| | - Hanan Nabat
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700, Ariel, Israel
| | - Nina Fainstein
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700, Ariel, Israel
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Efrat Mediouni
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700, Ariel, Israel
| | - Yarden Asis
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700, Ariel, Israel
| | - Paschalis Theotokis
- B' Department of Neurology, AHEPA University Hospital of Thessaloniki, Thessaloniki, Greece
| | - Ilias Salamotas
- B' Department of Neurology, AHEPA University Hospital of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- B' Department of Neurology, AHEPA University Hospital of Thessaloniki, Thessaloniki, Greece
| | - Abram Katz
- Åstrand Laboratory, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
| | - Tamir Ben-Hur
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Ofira Einstein
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700, Ariel, Israel.
| |
Collapse
|
4
|
Hamdi L, Nabat H, Goldberg Y, Fainstein N, Segal S, Mediouni E, Asis Y, Touloumi O, Grigoriadis N, Katz A, Ben-Hur T, Einstein O. Exercise training alters autoimmune cell invasion into the brain in autoimmune encephalomyelitis. Ann Clin Transl Neurol 2022; 9:1792-1806. [PMID: 36217574 DOI: 10.1002/acn3.51677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/01/2022] [Accepted: 09/23/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The mechanisms by which exercise training (ET) elicits beneficial effects on the systemic immune system and the central nervous system (CNS) in autoimmune neuroinflammation are not fully understood. OBJECTIVES To investigate (1) the systemic effects of high-intensity continuous training (HICT) on the migratory potential of autoimmune cells; (2) the direct effects of HICT on blood-brain-barrier (BBB) properties. METHODS Healthy mice were subjected to high-intensity continuous training (HICT) by treadmill running. The proteolipid protein (PLP) transfer EAE model was utilized to examine the immunomodulatory effects of training, where PLP-reactive lymph-node cells (LNCs) from HICT and sedentary donor mice were analyzed in vitro and transferred to naïve recipients that developed EAE. To examine neuroprotection, encephalitogenic LNCs from donor mice were transferred into HICT or sedentary recipient mice and the BBB was analyzed. RESULTS Transfer of PLP-reactive LNCs obtained from HICT donor mice attenuated EAE severity and inflammation in recipient mice. HICT markedly inhibited very late antigen (VLA)-4 and lymphocyte function-associated antigen (LFA)-1 expression in LNCs. Transfer of encephalitogenic LNCs into HICT recipients resulted in milder EAE and attenuated CNS inflammation. HICT reduced BBB permeability and the expression of intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1 in CNS blood vessels. INTERPRETATION HICT attenuates EAE development by both immunomodulatory and neuroprotective effects. The reduction in destructive CNS inflammation in EAE is attributed to systemic inhibition of autoreactive cell migratory potential, as well as reduction in BBB permeability, which are associated with reduced VLA-4/VCAM-1 and LFA-1/ICAM-1 interactions.
Collapse
Affiliation(s)
- Liel Hamdi
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | - Hanan Nabat
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | - Yehuda Goldberg
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | - Nina Fainstein
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Shir Segal
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | - Efrat Mediouni
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | - Yarden Asis
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | - Olga Touloumi
- B' Department of Neurology, AHEPA University Hospital of Thessaloniki, Greece
| | | | - Abram Katz
- Åstrand Laboratory, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
| | - Tamir Ben-Hur
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Ofira Einstein
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| |
Collapse
|
5
|
Bierhansl L, Hartung HP, Aktas O, Ruck T, Roden M, Meuth SG. Thinking outside the box: non-canonical targets in multiple sclerosis. Nat Rev Drug Discov 2022; 21:578-600. [PMID: 35668103 PMCID: PMC9169033 DOI: 10.1038/s41573-022-00477-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system that causes demyelination, axonal degeneration and astrogliosis, resulting in progressive neurological disability. Fuelled by an evolving understanding of MS immunopathogenesis, the range of available immunotherapies for clinical use has expanded over the past two decades. However, MS remains an incurable disease and even targeted immunotherapies often fail to control insidious disease progression, indicating the need for new and exceptional therapeutic options beyond the established immunological landscape. In this Review, we highlight such non-canonical targets in preclinical MS research with a focus on five highly promising areas: oligodendrocytes; the blood-brain barrier; metabolites and cellular metabolism; the coagulation system; and tolerance induction. Recent findings in these areas may guide the field towards novel targets for future therapeutic approaches in MS.
Collapse
Affiliation(s)
- Laura Bierhansl
- Department of Neurology, Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- German Center of Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
6
|
Chen J, Meng J, Li X, Li X, Liu Y, Jin C, Zhang L, Hao Z, Chen X, Zhang M, Liang C. HA/CD44 Regulates the T Helper 1 Cells Differentiation by Activating Annexin A1/Akt/mTOR Signaling to Drive the Pathogenesis of EAP. Front Immunol 2022; 13:875412. [PMID: 35693826 PMCID: PMC9178196 DOI: 10.3389/fimmu.2022.875412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
CD44 partcipates in multiple inflammatory reactions. Here, we aimed to investigate the role of CD44 and the ligand, hyaluronan (HA), on chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) pathogenesis. We found that CD44 was universally expressed in CD4+ lymphocytes in the peripheral blood of CP/CPPS patients. After silencing CD44 expression or delivering 4-methylumbelliferone (4-MU), the pain severity and prostatic inflammation were significantly relieved. In vitro assay found that HA/CD44 was able to regulate T helper 1 (Th1) cells differentiation, the deficiency of which diminished experimental autoimmune prostatitis (EAP) susceptibility. Bioinformatic analysis suggested that after HA or 4-MU treatment, mTOR signaling was significantly altered, and these results were confirmed by subsequent Western blotting assay. Besides, mass spectrometry and co-immunoprecipitation assays found that CD44 was able to interact with Annexin A1 (ANX A1), and this kind of interaction stabilized ANX A1 protein and maintained the activation of Akt/mTOR pathway. Meanwhile, HA-treatment-enhanced prostatic inflammation, Th1 cell differentiation, and Akt/mTOR pathway activation were reversed after silencing the expression of ANX A1 using shANX A1-lentivirus. The present study systematically investigates the functional role of HA/CD44 in CP/CPPS and identifies novel mechanisms for HA/CD44 promoting Th1 cell differentiation. Targeting the HA/CD44/ANX A1/Akt/mTOR signaling represents novel potential therapeutic strategies for patients with CP/CPPS.
Collapse
Affiliation(s)
- Jing Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Xiaoling Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Xiao Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Yi Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Chen Jin
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Zongyao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Xianguo Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- *Correspondence: Xianguo Chen, ; Meng Zhang, ; Chaozhao Liang,
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- *Correspondence: Xianguo Chen, ; Meng Zhang, ; Chaozhao Liang,
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- *Correspondence: Xianguo Chen, ; Meng Zhang, ; Chaozhao Liang,
| |
Collapse
|
7
|
Treatment of Experimental Autoimmune Encephalomyelitis with an Inhibitor of Phosphodiesterase-8 (PDE8). Cells 2022; 11:cells11040660. [PMID: 35203312 PMCID: PMC8870644 DOI: 10.3390/cells11040660] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
After decades of development, inhibitors targeting cyclic nucleotide phosphodiesterases (PDEs) expressed in leukocytes have entered clinical practice for the treatment of inflammatory disorders, with three PDE4 inhibitors being in clinical use as therapeutics for psoriasis, psoriatic arthritis, chronic obstructive pulmonary disease and atopic dermatitis. In contrast, the PDE8 family that is upregulated in pro-inflammatory T cells is a largely unexplored therapeutic target. We have previously demonstrated a role for the PDE8A-Raf-1 kinase complex in the regulation of myelin oligodendrocyte glycoprotein peptide 35–55 (MOG35–55) activated CD4+ effector T cell adhesion and locomotion by a mechanism that differs from PDE4 activity. In this study, we explored the in vivo treatment of experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis (MS) induced in mice immunized with MOG using the PDE8-selective inhibitor PF-04957325. For treatment in vivo, mice with EAE were either subcutaneously (s.c.) injected three times daily (10 mg/kg/dose), or were implanted subcutaneously with Alzet mini-osmotic pumps to deliver the PDE8 inhibitor (15.5 mg/kg/day). The mice were scored daily for clinical signs of paresis and paralysis which were characteristic of EAE. We observed the suppression of the clinical signs of EAE and a reduction of inflammatory lesion formation in the CNS by histopathological analysis through the determination of the numbers of mononuclear cells isolated from the spinal cord of mice with EAE. The PDE8 inhibitor treatment reduces the accumulation of both encephalitogenic Th1 and Th17 T cells in the CNS. Our study demonstrates the efficacy of targeting PDE8 as a treatment of autoimmune inflammation in vivo by reducing the inflammatory lesion load.
Collapse
|
8
|
Moser T, Hoepner L, Schwenker K, Seiberl M, Feige J, Akgün K, Haschke-Becher E, Ziemssen T, Sellner J. Cladribine Alters Immune Cell Surface Molecules for Adhesion and Costimulation: Further Insights to the Mode of Action in Multiple Sclerosis. Cells 2021; 10:cells10113116. [PMID: 34831335 PMCID: PMC8618022 DOI: 10.3390/cells10113116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/28/2022] Open
Abstract
Cladribine (CLAD) is a deoxyadenosine analogue prodrug which is given in multiple sclerosis (MS) as two short oral treatment courses 12 months apart. Reconstitution of adaptive immune function following selective immune cell depletion is the presumed mode of action. In this exploratory study, we investigated the impact of CLAD tablets on immune cell surface molecules for adhesion (CAMs) and costimulation (CoSs) in people with MS (pwMS). We studied 18 pwMS who started treatment with CLAD and 10 healthy controls (HCs). Peripheral blood mononuclear cells were collected at baseline and every 3 months throughout a 24-month period. We analysed ICAM-1, LFA-1, CD28, HLADR, CD154, CD44, VLA-4 (CD49d/CD29), PSGL-1 and PD-1 with regard to their expression on B and T cells (T helper (Th) and cytotoxic T cells (cT)) and surface density (mean fluorescence intensity, MFI) by flow cytometry. The targeted analysis of CAM and CoS on the surface of immune cells in pwMS revealed a higher percentage of ICAM-1 (B cells, Th, cT), LFA-1 (B cells, cT), HLADR (B cells, cT), CD28 (cT) and CD154 (Th). In pwMS, we found lower frequencies of Th and cT cells expressing PSGL-1 and B cells for the inhibitory signal PD-1, whereas the surface expression of LFA-1 on cT and of HLADR on B cells was denser. Twenty-four months after the first CLAD cycle, the frequencies of B cells expressing CD44, CD29 and CD49d were lower compared with the baseline, together with decreased densities of ICAM-1, CD44 and HLADR. The rate of CD154 expressing Th cells dropped at 12 months. For cT, no changes were seen for frequency or density. Immune reconstitution by oral CLAD was associated with modification of the pro-migratory and -inflammatory surface patterns of CAMs and CoSs in immune cell subsets. This observation pertains primarily to B cells, which are key cells underlying MS pathogenesis.
Collapse
Affiliation(s)
- Tobias Moser
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
- Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (L.H.); (K.A.); (T.Z.)
| | - Lena Hoepner
- Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (L.H.); (K.A.); (T.Z.)
| | - Kerstin Schwenker
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
| | - Michael Seiberl
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
| | - Julia Feige
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
| | - Katja Akgün
- Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (L.H.); (K.A.); (T.Z.)
| | | | - Tjalf Ziemssen
- Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, Carl Gustav Carus University Hospital, Technical University Dresden, 01307 Dresden, Germany; (L.H.); (K.A.); (T.Z.)
| | - Johann Sellner
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; (T.M.); (K.S.); (M.S.); (J.F.)
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, 80333 München, Germany
- Department of Neurology, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
- Correspondence: ; Tel.: +43-2572-9004-12850; Fax: +43-2572-9004-49281
| |
Collapse
|
9
|
Kotla NG, Bonam SR, Rasala S, Wankar J, Bohara RA, Bayry J, Rochev Y, Pandit A. Recent advances and prospects of hyaluronan as a multifunctional therapeutic system. J Control Release 2021; 336:598-620. [PMID: 34237401 DOI: 10.1016/j.jconrel.2021.07.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022]
Abstract
Hyaluronan (HA) is a naturally occurring non-sulfated glycosaminoglycan (GAG), cell-surface-associated biopolymer and is the key component of tissue extracellular matrix (ECM). Along with remarkable physicochemical properties, HA also has multifaceted biological effects that include but not limited to ECM organization, immunomodulation, and various cellular processes. Environmental cues such as tissue injury, infection or cancer change downstream signaling functionalities of HA. Unlike native HA, the fragments of HA have diversified effects on inflammation, cancer, fibrosis, angiogenesis and autoimmune response. In this review, we aim to discuss HA as a therapeutic delivery system development process, source, biophysical-chemical properties, and associated biological pathways (especially via cell surface receptors) of native and fragmented HA. We also tried to address an overview of the potential role of HA (native HA vs fragments) in the modulation of inflammation, immune response and various cancer targeting delivery applications. This review will also highlight the HA based therapeutic systems, medical devices and future perspectives of various biomedical applications were discussed in detail.
Collapse
Affiliation(s)
- Niranjan G Kotla
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, Paris F-75006, France
| | - Swetha Rasala
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Jitendra Wankar
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Raghvendra A Bohara
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, Paris F-75006, France; Indian Institute of Technology Palakkad, Palakkad 678 623, Kerala, India
| | - Yury Rochev
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland; Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, Moscow 119992, Russia.
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland.
| |
Collapse
|
10
|
Hamminger P, Marchetti L, Preglej T, Platzer R, Zhu C, Kamnev A, Rica R, Stolz V, Sandner L, Alteneder M, Kaba E, Waltenberger D, Huppa JB, Trauner M, Bock C, Lyck R, Bauer J, Dupré L, Seiser C, Boucheron N, Engelhardt B, Ellmeier W. Histone deacetylase 1 controls CD4 + T cell trafficking in autoinflammatory diseases. J Autoimmun 2021; 119:102610. [PMID: 33621930 DOI: 10.1016/j.jaut.2021.102610] [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: 12/13/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 01/13/2023]
Abstract
CD4+ T cell trafficking is a fundamental property of adaptive immunity. In this study, we uncover a novel role for histone deacetylase 1 (HDAC1) in controlling effector CD4+ T cell migration, thereby providing mechanistic insight into why a T cell-specific deletion of HDAC1 protects against experimental autoimmune encephalomyelitis (EAE). HDAC1-deficient CD4+ T cells downregulated genes associated with leukocyte extravasation. In vitro, HDAC1-deficient CD4+ T cells displayed aberrant morphology and migration on surfaces coated with integrin LFA-1 ligand ICAM-1 and showed an impaired ability to arrest on and to migrate across a monolayer of primary mouse brain microvascular endothelial cells under physiological flow. Moreover, HDAC1 deficiency reduced homing of CD4+ T cells into the intestinal epithelium and lamina propria preventing weight-loss, crypt damage and intestinal inflammation in adoptive CD4+ T cell transfer colitis. This correlated with reduced expression levels of LFA-1 integrin chains CD11a and CD18 as well as of selectin ligands CD43, CD44 and CD162 on transferred circulating HDAC1-deficient CD4+ T cells. Our data reveal that HDAC1 controls T cell-mediated autoimmunity via the regulation of CD4+ T cell trafficking into the CNS and intestinal tissues.
Collapse
MESH Headings
- Animals
- Autoimmunity
- Biomarkers
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Adhesion
- Chemotaxis, Leukocyte/genetics
- Chemotaxis, Leukocyte/immunology
- Disease Models, Animal
- Disease Susceptibility
- Encephalomyelitis, Autoimmune, Experimental/diagnosis
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Endothelial Cells
- Gene Expression Profiling
- Gene Expression Regulation
- Histone Deacetylase 1/genetics
- Histone Deacetylase 1/metabolism
- Immunohistochemistry
- Inflammation/diagnosis
- Inflammation/etiology
- Inflammation/metabolism
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice
- Mice, Knockout
Collapse
Affiliation(s)
- Patricia Hamminger
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Luca Marchetti
- Theodor Kocher Institute, University of Bern, Bern, Switzerland.
| | - Teresa Preglej
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria; Current Address: Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| | - René Platzer
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Ci Zhu
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria; Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| | - Anton Kamnev
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| | - Ramona Rica
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Valentina Stolz
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Lisa Sandner
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Marlis Alteneder
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Elisa Kaba
- Theodor Kocher Institute, University of Bern, Bern, Switzerland.
| | - Darina Waltenberger
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Johannes B Huppa
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Institute of Artificial Intelligence and Decision Support, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria.
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland.
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Austria.
| | - Loïc Dupré
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria; Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM UMR1291, CNRS UMR5051, Toulouse III Paul Sabatier University, Toulouse, France.
| | - Christian Seiser
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria.
| | - Nicole Boucheron
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | | | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
11
|
Epstein PM, Basole C, Brocke S. The Role of PDE8 in T Cell Recruitment and Function in Inflammation. Front Cell Dev Biol 2021; 9:636778. [PMID: 33937235 PMCID: PMC8085600 DOI: 10.3389/fcell.2021.636778] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/29/2021] [Indexed: 01/07/2023] Open
Abstract
Inhibitors targeting cyclic nucleotide phosphodiesterases (PDEs) expressed in leukocytes have entered clinical practice to treat inflammatory disorders, with three PDE4 inhibitors currently in clinical use as therapeutics for psoriasis, psoriatic arthritis, atopic dermatitis and chronic obstructive pulmonary disease. In contrast, the PDE8 family that is upregulated in pro-inflammatory T cells is a largely unexplored therapeutic target. It was shown that PDE8A plays a major role in controlling T cell and breast cancer cell motility, including adhesion to endothelial cells under physiological shear stress and chemotaxis. This is a unique function of PDE8 not shared by PDE4, another cAMP specific PDE, employed, as noted, as an anti-inflammatory therapeutic. Additionally, a regulatory role was shown for the PDE8A-rapidly accelerated fibrosarcoma (Raf)-1 kinase signaling complex in myelin antigen reactive CD4+ effector T cell adhesion and locomotion by a mechanism differing from that of PDE4. The PDE8A-Raf-1 kinase signaling complex affects T cell motility, at least in part, via regulating the LFA-1 integrin mediated adhesion to ICAM-1. The findings that PDE8A and its isoforms are expressed at higher levels in naive and myelin oligodendrocyte glycoprotein (MOG)35–55 activated effector T (Teff) cells compared to regulatory T (Treg) cells and that PDE8 inhibition specifically affects MOG35–55 activated Teff cell adhesion, indicates that PDE8A could represent a new beneficial target expressed in pathogenic Teff cells in CNS inflammation. The implications of this work for targeting PDE8 in inflammation will be discussed in this review.
Collapse
Affiliation(s)
- Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, CT, United States
| | - Chaitali Basole
- Department of Immunology, UConn Health, Farmington, CT, United States
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, CT, United States
| |
Collapse
|
12
|
Anti-inflammatory activity of CD44 antibodies in murine immune thrombocytopenia is mediated by Fcγ receptor inhibition. Blood 2021; 137:2114-2124. [PMID: 33662988 DOI: 10.1182/blood.2020009497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/11/2021] [Indexed: 02/03/2023] Open
Abstract
Monoclonal immunoglobulin G (IgG) antibodies to CD44 (anti-CD44) are anti-inflammatory in numerous murine autoimmune models, but the mechanisms are poorly understood. Anti-CD44 anti-inflammatory activity shows complete therapeutic concordance with IV immunoglobulin (IVIg) in treating autoimmune disease models, making anti-CD44 a potential IVIg alternative. In murine immune thrombocytopenia (ITP), there is no mechanistic explanation for anti-CD44 activity, although anti-CD44 ameliorates disease similarly to IVIg. Here, we demonstrate a novel anti-inflammatory mechanism of anti-CD44 that explains disease amelioration by anti-CD44 in murine ITP. Macrophages treated with anti-CD44 in vitro had dramatically suppressed phagocytosis through FcγRs in 2 separate systems of IgG-opsonized platelets and erythrocytes. Phagocytosis inhibition by anti-CD44 was mediated by blockade of the FcγR IgG binding site without changing surface FcγR expression. Anti-CD44 of different subclasses revealed that FcγR blockade was specific to receptors that could be engaged by the respective anti-CD44 subclass, and Fc-deactivated anti-CD44 variants lost all FcγR-inhibiting activity. In vivo, anti-CD44 functioned analogously in the murine passive ITP model and protected mice from ITP when thrombocytopenia was induced through an FcγR that could be engaged by the CD44 antibody's subclass. Consistent with FcγR blockade, Fc-deactivated variants of anti-CD44 were completely unable to ameliorate ITP. Together, anti-CD44 inhibits macrophage FcγR function and ameliorates ITP consistent with an FcγR blockade mechanism. Anti-CD44 is a potential IVIg alternative and may be of particular benefit in ITP because of the significant role that FcγRs play in human ITP pathophysiology.
Collapse
|
13
|
Antihuman CD44 antibody BJ18 inhibits platelet phagocytosis by correcting aberrant FcɣR expression and M1 polarization in immune thrombocytopenia. Int Immunopharmacol 2021; 95:107502. [PMID: 33690000 DOI: 10.1016/j.intimp.2021.107502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Immune thrombocytopenia (ITP) is an autoimmune hemorrhagic disease with a low platelet count. CD44 is a pivotal component involved in phagocytosis and inflammation, and monoclonal antibodies (mAbs) against CD44 have been shown to be beneficial in several autoimmune diseases. In the present study, we investigated the correlation between CD44 levels and disease severity in patients with ITP and explored the immunomodulatory mechanisms of the antihuman CD44 mAb BJ18 on platelet phagocytosis mediated by monocytes/macrophages. METHODS Plasma was collected from 45 participants to measure the circulating concentration of CD44 using ELISA. Peripheral blood mononuclear cells from patients and controls were isolated and induced to differentiate into monocytes/macrophages utilizing cytokines and drugs. CD44 expression on circulating cells and the effects of BJ18 on platelet phagocytosis, Fcɣ receptor (FcɣR) expression and M1/M2 polarization of macrophages were evaluated using flow cytometry and qPCR. RESULTS CD44 levels of both the soluble form found in plasma and the form expressed on the surface of circulating monocytes/macrophages were significantly elevated in ITP patients. Linear correlations were verified between the CD44 levels and major clinical characteristics. In an in vitro study, BJ18 successfully inhibited platelet phagocytosis by monocytes/macrophages obtained from ITP patients. Further studies indicated that BJ18 corrected abnormal FcγR expression on monocytes/macrophages. Moreover, the polarization of proinflammatory M1 macrophages could also be regulated by BJ18. CONCLUSIONS Our data indicated that the CD44 level has potential predictive value for disease severity and that the antihuman CD44 mAb BJ18 may be a promising therapy for ITP patients.
Collapse
|
14
|
Lind L, Svensson A, Thörn K, Krzyzowska M, Eriksson K. CD8 + T cells in the central nervous system of mice with herpes simplex infection are highly activated and express high levels of CCR5 and CXCR3. J Neurovirol 2021; 27:145-153. [PMID: 33492607 PMCID: PMC7831625 DOI: 10.1007/s13365-020-00940-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/09/2020] [Accepted: 12/27/2020] [Indexed: 12/25/2022]
Abstract
Herpes simplex virus type 2 (HSV-2) is a neurotropic virus that can cause meningitis, an inflammation of the meninges in the central nervous system. T cells are key players in viral clearance, and these cells migrate from peripheral blood into the central nervous system upon infection. Several factors contribute to T cell migration, including the expression of chemokines in the inflamed tissue that attract T cells through their expression of chemokine receptors. Here we investigated CD8+ T cell profile in the spinal cord in a mouse model of herpes simplex virus type 2 neuroinflammation. Mice were infected with HSV-2 and sacrificed when showing signs of neuroinflammation. Cells and/or tissue from spinal cord, spleen, and blood were analyzed for expression of activation markers, chemokine receptors, and chemokines. High numbers of CD8+ T cells were present in the spinal cord following genital HSV-2-infection. CD8+ T cells were highly activated and HSV-2 glycoprotein B -specific effector cells, some of which showed signs of recent degranulation. They also expressed high levels of many chemokine receptors, in particular CCR2, CCR4, CCR5, and CXCR3. Investigating corresponding receptor ligands in spinal cord tissue revealed markedly increased expression of the cognate ligands CCL2, CCL5, CCL8, CCL12, and CXCL10. This study shows that during herpesvirus neuroinflammation anti-viral CD8+ T cells accumulate in the CNS. CD8+ T cells in the CNS also express chemotactic receptors cognate to the chemotactic gradients in the spinal cord. This indicates that anti-viral CD8+ T cells may migrate to infected areas in the spinal cord during herpesvirus neuroinflammation in response to chemotactic gradients.
Collapse
Affiliation(s)
- Liza Lind
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alexandra Svensson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Karolina Thörn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Malgorzata Krzyzowska
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163, Warsaw, Poland
| | - Kristina Eriksson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
15
|
Agnew-Francis KA, Williams CM. Squaramides as Bioisosteres in Contemporary Drug Design. Chem Rev 2020; 120:11616-11650. [DOI: 10.1021/acs.chemrev.0c00416] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kylie A. Agnew-Francis
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Craig M. Williams
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
16
|
Reinbach C, Stadler MS, Pröbstl N, Chrzanowski U, Schmitz C, Kipp M, Hochstrasser T. CD44 expression in the cuprizone model. Brain Res 2020; 1745:146950. [PMID: 32524994 DOI: 10.1016/j.brainres.2020.146950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/08/2020] [Accepted: 06/05/2020] [Indexed: 02/03/2023]
Abstract
Numerous studies report that changes in extracellular matrix components and receptors, such as CD44, contribute to immune cell recruitment and thus lesion formation in multiple sclerosis (MS). In the present study, we used the cuprizone model to elucidate the expression pattern of CD44 in a toxin-induced MS model. Therefore, tissues of cuprizone-intoxicated mice were analyzed by real-time qRT-PCR and immunohistochemical staining against CD44. Co-localization analyses of CD44-positive cells with glial cell markers were performed by immunofluorescence labeling and in-situ hybridization. To investigate the functional importance of CD44 expression for myelination and glial cell activation, Cd44-deficient mice were used. In this study we demonstrate that CD44 expression is induced in a time-dependent manner in an autoimmune-independent model of MS. Up-regulation of CD44 expression was primarily associated to the superficial and perivascular glia limitans and demyelinated white matter structures, particularly the corpus callosum. In the demyelinated corpus callosum, CD44 was localized on GFAP+ astrocytes and IBA1+ microglial cells. Despite a robust expression induction, Cd44-deficiency did not ameliorate cuprizone-induced pathology. Although further studies will be needed to examine the functional relevance of CD44 in the cuprizone model, the spatial and temporal expression pattern of CD44 will pave the way to evaluate its precise role in different (immune and non-immune) pathological conditions.
Collapse
Affiliation(s)
- Christin Reinbach
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Maria-Sophia Stadler
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Nicolas Pröbstl
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Uta Chrzanowski
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Christoph Schmitz
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057 Rostock, Germany
| | - Tanja Hochstrasser
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Pettenkoferstr. 11, 80336 Munich, Germany.
| |
Collapse
|
17
|
Al-Ahmad AJ, Patel R, Palecek SP, Shusta EV. Hyaluronan impairs the barrier integrity of brain microvascular endothelial cells through a CD44-dependent pathway. J Cereb Blood Flow Metab 2019; 39:1759-1775. [PMID: 29589805 PMCID: PMC6727144 DOI: 10.1177/0271678x18767748] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hyaluronan (HA) constitutes the most abundant extracellular matrix component during brain development, only to become a minor component rapidly after birth and in adulthood to remain in specified regions. HA signaling has been associated with several neurological disorders, yet the impact of HA signaling at the blood-brain barrier (BBB) function remains undocumented. In this study, we investigated the impact of HA on BBB properties using human-induced pluripotent stem cell (iPSC) -derived and primary human and rat BMECs. The impact of HA signaling on developmental and mature BMECs was assessed by measuring changes in TEER, permeability, BMECs markers (GLUT1, tight junction proteins, P-gp) expression and localization, CD44 expression and hyaluronan levels. In general, HA treatment decreased barrier function and reduced P-gp activity with effects being more prominent upon treatment with oligomeric forms of HA (oHA). Such effects were exacerbated when applied during BMEC differentiation phase (considered as developmental BBB). We noted a hyaluronidase activity as well as an increase in CD44 expression during prolonged oxygen-glucose deprivation stress. Inhibition of HA signaling by antibody blockade of CD44 abrogated the detrimental effects of HA treatment. These results suggest the importance of HA signaling through CD44 on BBB properties.
Collapse
Affiliation(s)
- Abraham J Al-Ahmad
- 1 Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA.,2 Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Ronak Patel
- 2 Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Sean P Palecek
- 1 Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- 1 Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
18
|
Stephenson EL, Mishra MK, Moussienko D, Laflamme N, Rivest S, Ling CC, Yong VW. Chondroitin sulfate proteoglycans as novel drivers of leucocyte infiltration in multiple sclerosis. Brain 2019; 141:1094-1110. [PMID: 29506186 DOI: 10.1093/brain/awy033] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 01/04/2018] [Indexed: 12/28/2022] Open
Abstract
Multiple sclerosis presents with profound changes in the network of molecules involved in maintaining central nervous system architecture, the extracellular matrix. The extracellular matrix components, particularly the chondroitin sulfate proteoglycans, have functions beyond structural support including their potential interaction with, and regulation of, inflammatory molecules. To investigate the roles of chondroitin sulfate proteoglycans in multiple sclerosis, we used the experimental autoimmune encephalomyelitis model in a time course study. We found that the 4-sulfated glycosaminoglycan side chains of chondroitin sulfate proteoglycans, and the core protein of a particular family member, versican V1, were upregulated in the spinal cord of mice at peak clinical severity, correspondent with areas of inflammation. Versican V1 expression in the spinal cord rose progressively over the course of experimental autoimmune encephalomyelitis. A particular structure in the spinal cord and cerebellum that presented with intense upregulation of chondroitin sulfate proteoglycans is the leucocyte-containing perivascular cuff, an important portal of entry of immune cells into the central nervous system parenchyma. In these inflammatory perivascular cuffs, versican V1 and the glycosaminoglycan side chains of chondroitin sulfate proteoglycans were observed by immunohistochemistry within and in proximity to lymphocytes and macrophages as they migrated across the basement membrane into the central nervous system. Expression of versican V1 transcript was also documented in infiltrating CD45+ leucocytes and F4/80+ macrophages by in situ hybridization. To test the hypothesis that the chondroitin sulfate proteoglycans regulate leucocyte mobility, we used macrophages in tissue culture studies. Chondroitin sulfate proteoglycans significantly upregulated pro-inflammatory cytokines and chemokines in macrophages. Strikingly, and more potently than the toll-like receptor-4 ligand lipopolysaccharide, chondroitin sulfate proteoglycans increased the levels of several members of the matrix metalloproteinase family, which are implicated in the capacity of leucocytes to cross barriers. In support, the migratory capacity of macrophages in vitro in a Boyden chamber transwell assay was enhanced by chondroitin sulfate proteoglycans. Finally, using brain specimens from four subjects with multiple sclerosis with active lesions, we found chondroitin sulfate proteoglycans to be associated with leucocytes in inflammatory perivascular cuffs in all four patients. We conclude that the accumulation of chondroitin sulfate proteoglycans in the perivascular cuff in multiple sclerosis and experimental autoimmune encephalomyelitis boosts the activity and migration of leucocytes across the glia limitans into the central nervous system parenchyma. Thus, chondroitin sulfate proteoglycans represent a new class of molecules to overcome in order to reduce the inflammatory cascades and clinical severity of multiple sclerosis.
Collapse
Affiliation(s)
- Erin L Stephenson
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Manoj K Mishra
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Daniel Moussienko
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Nataly Laflamme
- Department of Molecular Medicine, CHU de Quebec Research Center, Laval University, Quebec, Canada
| | - Serge Rivest
- Department of Molecular Medicine, CHU de Quebec Research Center, Laval University, Quebec, Canada
| | - Chang-Chun Ling
- Department of Chemistry, University of Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| |
Collapse
|
19
|
Lim HW, Pak K, Kurabi A, Ryan AF. Lack of the hyaluronan receptor CD44 affects the course of bacterial otitis media and reduces leukocyte recruitment to the middle ear. BMC Immunol 2019; 20:20. [PMID: 31226944 PMCID: PMC6588864 DOI: 10.1186/s12865-019-0302-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 06/09/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND CD44 is a multifunctional molecule that plays major roles in both leukocyte recruitment and tissue proliferation. Since mucosal hyperplasia and leukocyte infiltration of the middle ear cavity are major features of otitis media, we evaluated the role of CD44 in the pathophysiology and course of this disease in a mouse model of middle ear infection. Expression of genes related to CD44 function were evaluated using gene arrays in wild-type mice. The middle ears of mice deficient in CD44 were inoculated with non-typeable Haemophilus influenzae. Histopathology and bacterial clearance were compared to that seen in wild-type controls. RESULTS We observed strong up-regulation of CD44 and of genes related to its role in leukocyte extravasation into the middle ear, during the course of acute otitis media. Mice deficient in CD44 exhibited reduced early mucosal hyperplasia and leukocyte recruitment, followed by delayed resolution of infection and persistent inflammation. CONCLUSIONS CD44 plays an important role in OM pathogenesis by altering the mucosal growth and neutrophil enlistment. Targeted therapies based on CD44 could be useful adjuncts to the treatment of middle ear infections.
Collapse
Affiliation(s)
- Hyun Woo Lim
- Department of Surgery/Otolaryngology, University of California-San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093-0666 USA
- Department of Otolaryngology, University of Ulsan College of Medicine, Gangneung, South Korea
| | - Kwang Pak
- Department of Surgery/Otolaryngology, University of California-San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093-0666 USA
| | - Arwa Kurabi
- Department of Surgery/Otolaryngology, University of California-San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093-0666 USA
| | - Allen F. Ryan
- Department of Surgery/Otolaryngology, University of California-San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093-0666 USA
- San Diego VA Medical Center, La Jolla, CA USA
| |
Collapse
|
20
|
Medina CO, Nagy N, Bollyky PL. Extracellular matrix and the maintenance and loss of peripheral immune tolerance in autoimmune insulitis. Curr Opin Immunol 2018; 55:22-30. [PMID: 30248522 DOI: 10.1016/j.coi.2018.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022]
Abstract
There is a growing appreciation that the extracellular matrix (ECM) contributes to both the maintenance of immune tolerance in healthy tissues and to its loss at sites of autoimmunity. Here, we review recent literature on the role of ECM and particularly the glycosaminoglycans hyaluronan and heparan sulfate in the development of autoimmune, type 1 diabetes (T1D). Data from transplant models suggest that healthy islets are embedded within an intact ECM that supports beta-cell homeostasis and provides physical and immunoregulatory barriers against immune infiltration. However, studies of human insulitis as well as the non-obese diabetic (NOD) and DORmO mouse models of T1D indicate that autoimmune insulitis is associated with the degradation of basement membrane structures, the catabolism of the islet interstitium, and the accumulation of a hyaluronan-rich, pro-inflammatory ECM. Moreover, in these models of autoimmune diabetes, either the pharmacologic inhibition of heparan sulfate catabolism, the reduction of hyaluronan synthesis, or the targeting of the pathways that sense these ECM changes can all prevent beta-cell destruction. Together these data support an emerging paradigm that in healthy islets the local ECM contributes to both immune tolerance and beta-cell homeostasis while in chronic inflammation the islet ECM is permissive to immune infiltration and beta-cell destruction. Therapies that support ECM-mediated 'barrier tolerance' may have potential as adjunctive agents in combination regimens designed to prevent or treat autoimmunity.
Collapse
Affiliation(s)
- Carlos O Medina
- Division of Infectious Diseases and Geographic Medicine, Dept. of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA, 94305, United States
| | - Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Dept. of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA, 94305, United States
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Dept. of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA, 94305, United States.
| |
Collapse
|
21
|
Stephenson EL, Yong VW. Pro-inflammatory roles of chondroitin sulfate proteoglycans in disorders of the central nervous system. Matrix Biol 2018; 71-72:432-442. [PMID: 29702175 DOI: 10.1016/j.matbio.2018.04.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/21/2018] [Accepted: 04/21/2018] [Indexed: 02/06/2023]
Abstract
The extracellular matrix of the central nervous system is an interconnected network of proteins and sugars. It is crucial for homeostasis, but its remodeling in neurological diseases impacts both injury and repair. Here we introduce an extracellular matrix family member that participates in immune-matrix interactions, the chondroitin sulfate proteoglycans. Chondroitin sulfate proteoglycans integrate signals from the microenvironment to activate immune cells, and they boost inflammatory responses by binding immunological receptors including toll-like receptors, selectins, CD44, and β1 integrin. Chondroitin sulfate proteoglycans also bind signaling molecules for immune cells such as cytokines and chemokines, and they activate matrix-degrading enzymes. Chondroitin sulfate proteoglycans accumulate in the damaged CNS, including during traumatic brain/spinal cord injury and multiple sclerosis, and they help drive pathogenesis. This Review aims to give new insights into the remodeling of chondroitin sulfate proteoglycans during inflammation, and how these matrix glycoproteins are able to drive neuroinflammation.
Collapse
Affiliation(s)
- Erin L Stephenson
- Hotchkiss Brain Institute and the University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
22
|
Nagy N, Kuipers HF, Marshall PL, Wang E, Kaber G, Bollyky PL. Hyaluronan in immune dysregulation and autoimmune diseases. Matrix Biol 2018; 78-79:292-313. [PMID: 29625181 DOI: 10.1016/j.matbio.2018.03.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/10/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Abstract
The tissue microenvironment contributes to local immunity and to the pathogenesis of autoimmune diseases - a diverse set of conditions characterized by sterile inflammation, immunity against self-antigens, and destruction of tissues. However, the specific factors within the tissue microenvironment that contribute to local immune dysregulation in autoimmunity are poorly understood. One particular tissue component implicated in multiple autoimmune diseases is hyaluronan (HA), an extracellular matrix (ECM) polymer. HA is abundant in settings of chronic inflammation and contributes to lymphocyte activation, polarization, and migration. Here, we first describe what is known about the size, amount, and distribution of HA at sites of autoimmunity and in associated lymphoid structures in type 1 diabetes, multiple sclerosis, and rheumatoid arthritis. Next, we examine the recent literature on HA and its impact on adaptive immunity, particularly in regards to the biology of lymphocytes and Foxp3+ regulatory T-cells (Treg), a T-cell subset that maintains immune tolerance in healthy individuals. We propose that HA accumulation at sites of chronic inflammation creates a permissive environment for autoimmunity, characterized by CD44-mediated inhibition of Treg expansion. Finally, we address potential tools and strategies for targeting HA and its receptor CD44 in chronic inflammation and autoimmunity.
Collapse
Affiliation(s)
- Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Hedwich F Kuipers
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton L Marshall
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Esther Wang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
23
|
Blank T, Goldmann T, Koch M, Amann L, Schön C, Bonin M, Pang S, Prinz M, Burnet M, Wagner JE, Biel M, Michalakis S. Early Microglia Activation Precedes Photoreceptor Degeneration in a Mouse Model of CNGB1-Linked Retinitis Pigmentosa. Front Immunol 2018; 8:1930. [PMID: 29354133 PMCID: PMC5760536 DOI: 10.3389/fimmu.2017.01930] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/15/2017] [Indexed: 01/12/2023] Open
Abstract
Retinitis pigmentosa (RP) denotes a family of inherited blinding eye diseases characterized by progressive degeneration of rod and cone photoreceptors in the retina. In most cases, a rod-specific genetic defect results in early functional loss and degeneration of rods, which is followed by degeneration of cones and loss of daylight vision at later stages. Microglial cells, the immune cells of the central nervous system, are activated in retinas of RP patients and in several RP mouse models. However, it is still a matter of debate whether activated microglial cells may be responsible for the amplification of the typical degenerative processes. Here, we used Cngb1−/− mice, which represent a slow degenerative mouse model of RP, to investigate the extent of microglia activation in retinal degeneration. With a combination of FACS analysis, immunohistochemistry and gene expression analysis we established that microglia in the Cngb1−/− retina were already activated in an early, predegenerative stage of the disease. The evidence available so far suggests that early retinal microglia activation represents a first step in RP, which might initiate or accelerate photoreceptor degeneration.
Collapse
Affiliation(s)
- Thomas Blank
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Goldmann
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,In Vivo Pharmacology, Synovo GmbH, Tübingen, Germany
| | - Mirja Koch
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lukas Amann
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Christian Schön
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Bonin
- Institute for Medical Genetics and Applied Genomics Transcriptomics, University of Tübingen, Tübingen, Germany.,IMGM Laboratories GmbH, Planegg, Germany
| | - Shengru Pang
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | | | - Johanna E Wagner
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Biel
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
24
|
Basole CP, Nguyen RK, Lamothe K, Vang A, Clark R, Baillie GS, Epstein PM, Brocke S. PDE8 controls CD4 + T cell motility through the PDE8A-Raf-1 kinase signaling complex. Cell Signal 2017; 40:62-72. [PMID: 28851628 DOI: 10.1016/j.cellsig.2017.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 08/01/2017] [Accepted: 08/23/2017] [Indexed: 11/19/2022]
Abstract
The levels of cAMP are regulated by phosphodiesterase enzymes (PDEs), which are targets for the treatment of inflammatory disorders. We have previously shown that PDE8 regulates T cell motility. Here, for the first time, we report that PDE8A exerts part of its control of T cell function through the V-raf-1 murine leukemia viral oncogene homolog 1 (Raf-1) kinase signaling pathway. To examine T cell motility under physiologic conditions, we analyzed T cell interactions with endothelial cells and ligands in flow assays. The highly PDE8-selective enzymatic inhibitor PF-04957325 suppresses adhesion of in vivo myelin oligodendrocyte glycoprotein (MOG) activated inflammatory CD4+ T effector (Teff) cells to brain endothelial cells under shear stress. Recently, PDE8A was shown to associate with Raf-1 creating a compartment of low cAMP levels around Raf-1 thereby protecting it from protein kinase A (PKA) mediated inhibitory phosphorylation. To test the function of this complex in Teff cells, we used a cell permeable peptide that selectively disrupts the PDE8A-Raf-1 interaction. The disruptor peptide inhibits the Teff-endothelial cell interaction more potently than the enzymatic inhibitor. Furthermore, the LFA-1/ICAM-1 interaction was identified as a target of disruptor peptide mediated reduction of adhesion, spreading and locomotion of Teff cells under flow. Mechanistically, we observed that disruption of the PDE8A-Raf-1 complex profoundly alters Raf-1 signaling in Teff cells. Collectively, our studies demonstrate that PDE8A inhibition by enzymatic inhibitors or PDE8A-Raf-1 kinase complex disruptors decreases Teff cell adhesion and migration under flow, and represents a novel approach to target T cells in inflammation.
Collapse
Affiliation(s)
| | | | - Katie Lamothe
- Department of Immunology, UConn Health, United States
| | - Amanda Vang
- Department of Immunology, UConn Health, United States; The National Hospital of Faroe Islands, Faroe Islands
| | - Robert Clark
- Department of Immunology, UConn Health, United States
| | - George S Baillie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | | | - Stefan Brocke
- Department of Immunology, UConn Health, United States.
| |
Collapse
|
25
|
Chrysin Attenuates VCAM-1 Expression and Monocyte Adhesion in Lipopolysaccharide-Stimulated Brain Endothelial Cells by Preventing NF-κB Signaling. Int J Mol Sci 2017; 18:ijms18071424. [PMID: 28671640 PMCID: PMC5535915 DOI: 10.3390/ijms18071424] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/22/2017] [Accepted: 06/27/2017] [Indexed: 02/07/2023] Open
Abstract
Adhesion of leukocytes to endothelial cells plays an important role in neuroinflammation. Therefore, suppression of the expression of adhesion molecules in brain endothelial cells may inhibit neuroinflammation. Chrysin (5,7-dihydroxyflavone) is a flavonoid component of propolis, blue passion flowers, and fruits. In the present study, we examined the effects of chrysin on lipopolysaccharide (LPS)-induced expression of vascular cell adhesion molecule-1 (VCAM-1) in mouse cerebral vascular endothelial (bEnd.3) cells. In bEnd.3 cells, LPS increased mRNA expression of VCAM-1 in a time-dependent manner, and chrysin significantly decreased LPS-induced mRNA expression of VCAM-1. Chrysin also reduced VCAM-1 protein expression in a concentration-dependent manner. Furthermore, chrysin blocked adhesion of monocytes to bEnd.3 cells exposed to LPS. Nuclear factor-κB (NF-κB), p38 mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinase, which are all activated by LPS, were significantly inhibited by chrysin. These results indicate that chrysin inhibits the expression of VCAM-1 in brain endothelial cells by inhibiting NF-κB translocation and MAPK signaling, resulting in the attenuation of leukocyte adhesion to endothelial cells. The anti-inflammatory effects of chrysin suggest a possible therapeutic application of this agent to neurodegenerative diseases, such as multiple sclerosis, septic encephalopathy, and allergic encephalomyelitis.
Collapse
|
26
|
Pinner E, Gruper Y, Ben Zimra M, Kristt D, Laudon M, Naor D, Zisapel N. CD44 Splice Variants as Potential Players in Alzheimer’s Disease Pathology. J Alzheimers Dis 2017; 58:1137-1149. [DOI: 10.3233/jad-161245] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | | | - Micha Ben Zimra
- The Lautenberg Center for General and Tumor Immunology, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Don Kristt
- Molecular Pathology Unit, Rabin Medical Center, Petah Tikva, Israel
| | | | - David Naor
- The Lautenberg Center for General and Tumor Immunology, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Nava Zisapel
- Neurim Pharmaceuticals Ltd, Tel-Aviv, Israel
- Department of Neurobiology Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
27
|
Chitrala KN, Guan H, Singh NP, Busbee B, Gandy A, Mehrpouya-Bahrami P, Ganewatta MS, Tang C, Chatterjee S, Nagarkatti P, Nagarkatti M. CD44 deletion leading to attenuation of experimental autoimmune encephalomyelitis results from alterations in gut microbiome in mice. Eur J Immunol 2017; 47:1188-1199. [PMID: 28543188 DOI: 10.1002/eji.201646792] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/17/2017] [Accepted: 05/10/2017] [Indexed: 02/06/2023]
Abstract
Dysbiosis in gut microbiome has been shown to be associated with inflammatory and autoimmune diseases. Previous studies from our laboratory demonstrated the pivotal role played by CD44 in the regulation of EAE, a murine model of multiple sclerosis. In the current study, we determined whether these effects resulted from an alteration in gut microbiota and the short-chain fatty acid (SCFA) production in CD44 knockout (CD44KO) mice. Fecal transfer from naïve CD44KO but not C57BL/6 wild type (CD44WT) mice, into EAE-induced CD44WT mice, led to significant amelioration of EAE. High-throughput bacterial 16S rRNA gene sequencing, followed by clustering sequences into operational taxonomic units (OTUs) and biochemical analysis, revealed that EAE-induced CD44KO mice showed significant diversity, richness, and evenness when compared to EAE-induced CD44WT mice at the phylum level, with dominant Bacteroidetes (68.5%) and low Firmicutes (26.8%). Further, data showed a significant change in the abundance of SCFAs, propionic acid, and i-butyric acid in EAE-CD44KO compared to EAE-CD44WT mice. In conclusion, our results demonstrate that the attenuation of EAE seen following CD44 gene deletion in mice may result from alterations in the gut microbiota and SCFAs. Furthermore, our studies also demonstrate that the phenotype of gene knock-out animals may be shaped by gut microbiota.
Collapse
Affiliation(s)
| | - Hongbing Guan
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, Columbia, SC, USA.,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Narendra P Singh
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, Columbia, SC, USA
| | - Brandon Busbee
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, Columbia, SC, USA
| | - Alexa Gandy
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, Columbia, SC, USA
| | - Pegah Mehrpouya-Bahrami
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, Columbia, SC, USA
| | - Mitra S Ganewatta
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Chuanbing Tang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Saurabh Chatterjee
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, Columbia, SC, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, Columbia, SC, USA.,WJB Dorn VA Medical Center, Columbia, SC, USA
| |
Collapse
|
28
|
Sagar D, Singh NP, Ginwala R, Huang X, Philip R, Nagarkatti M, Nagarkatti P, Neumann K, Ruland J, Andrews AM, Ramirez SH, Khan ZK, Jain P. Antibody blockade of CLEC12A delays EAE onset and attenuates disease severity by impairing myeloid cell CNS infiltration and restoring positive immunity. Sci Rep 2017; 7:2707. [PMID: 28578388 PMCID: PMC5457463 DOI: 10.1038/s41598-017-03027-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/03/2017] [Indexed: 12/21/2022] Open
Abstract
The mechanism of dendritic cells (DCs) recruitment across the blood brain barrier (BBB) during neuroinflammation has been the least explored amongst all leukocytes. For cells of myeloid origin, while integrins function at the level of adhesion, the importance of lectins remains unknown. Here, we identified functions of one C-type lectin receptor, CLEC12A, in facilitating DC binding and transmigration across the BBB in response to CCL2 chemotaxis. To test function of CLEC12A in an animal model of multiple sclerosis (MS), we administered blocking antibody to CLEC12A that significantly ameliorated disease scores in MOG35–55-induced progressive, as well as PLP138–151-induced relapsing-remitting experimental autoimmune encephalomyelitis (EAE) mice. The decline in both progression and relapse of EAE occurred as a result of reduced demyelination and myeloid cell infiltration into the CNS tissue. DC numbers were restored in the spleen of C57BL/6 and peripheral blood of SJL/J mice along with a decreased TH17 phenotype within CD4+ T-cells. The effects of CLEC12A blocking were further validated using CLEC12A knockout (KO) animals wherein EAE disease induction was delayed and reduced disease severity was observed. These studies reveal the utility of a DC-specific mechanism in designing new therapeutics for MS.
Collapse
Affiliation(s)
- Divya Sagar
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Narendra P Singh
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Rashida Ginwala
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Xiaofang Huang
- Immunotope Inc., Pennsylvania Biotechnology Center, Doylestown, PA, USA
| | - Ramila Philip
- Immunotope Inc., Pennsylvania Biotechnology Center, Doylestown, PA, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA.,William Jennings Bryan Dorn VA Medical Center, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Konstantin Neumann
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Allison M Andrews
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Servio H Ramirez
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Zafar K Khan
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Pooja Jain
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Karta MR, Rosenthal PS, Beppu A, Vuong CY, Miller M, Das S, Kurten RC, Doherty TA, Broide DH. β 2 integrins rather than β 1 integrins mediate Alternaria-induced group 2 innate lymphoid cell trafficking to the lung. J Allergy Clin Immunol 2017; 141:329-338.e12. [PMID: 28366795 DOI: 10.1016/j.jaci.2017.03.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 01/23/2017] [Accepted: 03/15/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2s) expand in the lungs of mice during type 2 inflammation induced by the fungal allergen Alternaria alternata. The increase in ILC2 numbers in the lung has been largely attributed to local proliferation and whether ILC2s migrate from the circulation to the lung after Alternaria exposure is unknown. OBJECTIVE We examined whether human (lung, lymph node, and blood) and mouse lung ILC2s express β1 and β2 integrin adhesion molecules and whether these integrins are required for trafficking of ILC2s into the lungs of mice. METHODS Human and mouse ILC2s were assessed for surface expression of β1 and β2 integrin adhesion molecules by using flow cytometry. The role of β1 and β2 integrins in ILC2 trafficking to the lungs was assessed by in vivo blocking of these integrins before airway exposure to Alternaria in mice. RESULTS Both human and mouse lung ILC2s express high levels of β1 and β2 integrin adhesion receptors. Intranasal administration of Alternaria challenge reduced ILC2 numbers in the bone marrow and concurrently increased blood and lung ILC2 numbers. In vivo blocking of β2 integrins (CD18) significantly reduced ILC2 numbers in the lungs but did not alter ILC2 proliferation, apoptosis, and function. In contrast, in vivo blocking of β1 integrins or α4 integrins did not affect lung ILC2 numbers. CONCLUSION ILC2 numbers increase in the mouse lung not only through local proliferation but also through trafficking from the circulation into the lung using β2 rather than β1 or α4 integrins.
Collapse
Affiliation(s)
- Maya R Karta
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Peter S Rosenthal
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Andrew Beppu
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Christine Y Vuong
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Marina Miller
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Sudipta Das
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Richard C Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital Research Institute, Little Rock, Ark
| | - Taylor A Doherty
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - David H Broide
- Department of Medicine, University of California San Diego, La Jolla, Calif.
| |
Collapse
|
30
|
Franck S, Paterka M, Birkenstock J, Zipp F, Siffrin V, Witsch E. Phenotype of Antigen Unexperienced T H Cells in the Inflamed Central Nervous System in Experimental Autoimmune Encephalomyelitis. J Neuroimmune Pharmacol 2016; 12:305-313. [PMID: 27832402 DOI: 10.1007/s11481-016-9718-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/28/2016] [Indexed: 10/20/2022]
Abstract
Multiple sclerosis is a chronic, disseminated inflammation of the central nervous system which is thought to be driven by autoimmune T cells. Genetic association studies in multiple sclerosis and a large number of studies in the animal model of the disease support a role for effector/memory T helper cells. However, the mechanisms underlying relapses, remission and chronic progression in multiple sclerosis or the animal model experimental autoimmune encephalomyelitis, are not clear. In particular, there is only scarce information on the role of central nervous system-invading naive T helper cells in these processes. By applying two-photon laser scanning microscopy we could show in vivo that antigen unexperienced T helper cells migrated into the deep parenchyma of the inflamed central nervous system in experimental autoimmune encephalomyelitis, independent of their antigen specificity. Using flow cytometric analyses of central nervous system-derived lymphocytes we found that only antigen-specific, formerly naive T helper cells became activated during inflammation of the central nervous system encountering their corresponding antigen.
Collapse
Affiliation(s)
- Sophia Franck
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Magdalena Paterka
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Jerome Birkenstock
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Volker Siffrin
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Esther Witsch
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany.
| |
Collapse
|
31
|
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]
|
32
|
Anstadt EJ, Fujiwara M, Wasko N, Nichols F, Clark RB. TLR Tolerance as a Treatment for Central Nervous System Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2016; 197:2110-8. [PMID: 27503211 DOI: 10.4049/jimmunol.1600876] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/10/2016] [Indexed: 12/16/2022]
Abstract
The role of TLR signaling in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) is unclear. This role is especially controversial in models of adoptive transfer EAE in which no adjuvant and no TLR ligands are administered. We recently reported that a microbiome-derived TLR2 ligand, Lipid 654 (L654), is present in healthy human serum but significantly decreased in the serum of MS patients. This suggested that microbiome products that gain access to the systemic circulation, rather than being proinflammatory, may normally play an immune-regulatory role by maintaining a state of relative TLR tolerance. Therefore, a loss of microbiome-mediated TLR tolerance, as suggested by lower serum levels of L654, may play a role in the pathogenesis of MS. As proof of concept we asked whether administering low-level TLR2 ligands in adoptive transfer EAE induces TLR2 tolerance and attenuates disease. We administered low-level Pam2CSK4 or L654 to mice receiving encephalitogenic cells and in doing so induced both TLR2 tolerance and attenuation of EAE. Disease attenuation was accompanied in the CNS by a decrease in macrophage activation, a decrease in a specific proinflammatory macrophage population, and a decrease in Th17 cells. In addition, disease attenuation was associated with an increase in splenic type 1 regulatory T cells. Kinetic tolerance induction studies revealed a critical period for TLR2 involvement in adoptive transfer EAE. Overall, these results suggest that inducing TLR tolerance may offer a new approach to treating CNS autoimmune diseases such as MS.
Collapse
Affiliation(s)
- Emily J Anstadt
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06032; and
| | - Mai Fujiwara
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06032; and
| | - Nicholas Wasko
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06032; and
| | - Frank Nichols
- School of Dental Medicine, University of Connecticut School of Medicine, Farmington, CT 06032
| | - Robert B Clark
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06032; and
| |
Collapse
|
33
|
Piraino PS, Yednock TA, Freedman SB, Messersmith EK, Pleiss MA, Karlik SJ. Suppression of acute experimental allergic encephalomyelitis with a small molecule inhibitor of α4 integrin. Mult Scler 2016; 11:683-90. [PMID: 16320728 DOI: 10.1191/1352458505ms1223oa] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Purpose: To determine the efficacy of a small molecule inhibitor of α4 integrin (CT301) at reversing the clinical, pathological and MR- detectable deficits associated with the acute phase of experimental allergic encephalomyelitis (EAE). Materials and methods: EAE was induced in 36 female Hartley guinea pigs, and the treatment period was from day 11 to day 17 post-immunization. Animals received either saline (n=12), anti-α4 integrin antibody (AN100226m; n=12) or CT301 (n=12). T2-weighted fast spin echo and T1-weighted pre- and post-contrast scans were performed at the beginning (day 11) and end (day 18) of the treatment period, and scored for cerebral inflammation and gadolinium enhancement. T1-weighted images were further analyzed to quantify this enhancement as a measure of blood-brain barrier integrity. Dissected CNS was evaluated for inflammation and demyelination. Results: CT301 successfully reversed two clinical indicators of disease over the course of the treatment period. These animals showed decreased T2-weighted abnormalities, as well as a reduction in gadolinium leakage on T1-weighted images. Meningeal and perivascular inflammation was decreased by anti-α4 integrin treatments. Conclusion: CT301 effectively reverses the clinical, pathological and MR-detectable deficits of acute EAE, and may therefore be a promising therapeutic agent in multiple sclerosis (MS).
Collapse
Affiliation(s)
- P S Piraino
- Department of Physiology and Pharmacology, University of Western Ontario, London, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Ni X, Geller EB, Eppihimer MJ, Eisenstein TK, Adler MW, Tuma RF. Win 55212-2, a cannabinoid receptor agonist, attenuates leukocyte/endothelial interactions in an experimental autoimmune encephalomyelitis model. Mult Scler 2016; 10:158-64. [PMID: 15124761 DOI: 10.1191/1352458504ms1009oa] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Multiple sclerosis (MS) is the most common of the immune demyelinating disorders of the central nervous system (C NS). Leukocyte/endothelial interactions are important steps in the progression of the disease and substances that interfere with these activities have been evaluated as potential therapeutic agents. C annabinoid receptor agonists have been shown to downregulate immune responses and there is preliminary evidence that they may slow the progress of MS. The purpo se of this investigation was to determine how cannabinoid recepto r agonists interfere with leukocyte rolling and adhesion. This was investigated in an experimental autoimmune encephalo myelitis (EAE) model using six to eight week old C 57BL/6 mice. Mouse myelin oligodendrocyte protein and pertussis toxin were used to induce EAE. WIN 55212-2, C B1 and C B2 antagonist were given. By use of in vivo intravital microscopy, leukocyte/endothelial interactio ns were evaluated via a cranial window implanted two days before. The results demonstrated that EAE increases leukocyte rolling and firm adhesion in the brain, and that this increased leukocyte/endothelial interactio n can be attenuated by administration of WIN 55212-2. Furthermore, use of the selective antagonists for the C B1 recepto r (SR 141716A) and the C B2 receptor (SR144528) in this study demonstrated that the cannabinoid’s inhibitory effects on leukocyte/endothelial interactions can be mediated by activating C B2 receptor.
Collapse
MESH Headings
- Animals
- Benzoxazines
- Calcium Channel Blockers/pharmacology
- Cell Adhesion/drug effects
- Cell Adhesion/immunology
- Cell Communication/drug effects
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Endothelium, Vascular/cytology
- Female
- Leukocyte Rolling/drug effects
- Leukocytes/cytology
- Mice
- Mice, Inbred C57BL
- Morpholines/pharmacology
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Naphthalenes/pharmacology
- Paralysis/drug therapy
- Paralysis/immunology
- Paralysis/pathology
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB2/agonists
Collapse
Affiliation(s)
- Xiang Ni
- Department of Physiology, Temple University, 3420 N. Broad Street, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|
35
|
Steinman L. A Journey in Science: The Privilege of Exploring the Brain and the Immune System. Mol Med 2016; 22:molmed.2015.00263. [PMID: 27652378 PMCID: PMC5004718 DOI: 10.2119/molmed.2015.00263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/06/2022] Open
Abstract
Real innovations in medicine and science are historic and singular; the stories behind each occurrence are precious. At Molecular Medicine we have established the Anthony Cerami Award in Translational Medicine to document and preserve these histories. The monographs recount the seminal events as told in the voice of the original investigators who provided the crucial early insight. These essays capture the essence of discovery, chronicling the birth of ideas that created new fields of research; and launched trajectories that persisted and ultimately influenced how disease is prevented, diagnosed, and treated. In this volume, the Cerami Award Monograph is by Lawrence Steinman, MD, of Stanford University in California. A visionary in the field of neurology, this is the story of Dr. Steinman's scientific journey.
Collapse
Affiliation(s)
- Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California
| |
Collapse
|
36
|
Harris N, Koppel J, Zsila F, Juhas S, Il'kova G, Kogan FY, Lahmy O, Wildbaum G, Karin N, Zhuk R, Gregor P. Mechanism of action and efficacy of RX-111, a thieno[2,3-c]pyridine derivative and small molecule inhibitor of protein interaction with glycosaminoglycans (SMIGs), in delayed-type hypersensitivity, TNBS-induced colitis and experimental autoimmune encephalomyelitis. Inflamm Res 2016; 65:285-94. [PMID: 26794621 DOI: 10.1007/s00011-016-0915-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/04/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE AND DESIGN Elucidate the mechanism of action of the small molecule inhibitor of protein binding to glycosaminoglycans, RX-111 and assay its anti-inflammatory activity in animal models of inflammatory disease. MATERIALS The glycosaminoglycan, heparin, was used in the mechanism of action study of RX-111. Human T lymphocytes and umbilical vein endothelial cells were used to assay the in vitro activity of RX-111. Mouse and rat models of disease were used to assay the anti-inflammatory activity of RX-111 in vivo. METHODS Circular dichroism and UV/Vis absorption spectroscopy were used to study the binding of RX-111 to the glycosaminoglycan, heparin. T lymphocyte rolling on endothelial cells under shear flow was used to assay RX-111 activity in vitro. Delayed-type hypersensitivity (DTH) and tri-nitrobenzene sulfonic acid (TNBS)-induced colitis in mice and experimental autoimmune encephalomyelitis (EAE) in rats were used to assay anti-inflammatory activity of RX-111 in vivo. RESULTS RX-111 was shown to bind directly to heparin. It inhibited leukocyte rolling on endothelial cells under shear flow and reduced inflammation in the mouse model of DTH. RX-111 was efficacious in the mouse model of inflammatory bowel disease, TNBS-induced colitis and the rat model of multiple sclerosis, EAE. CONCLUSIONS RX-111 exercises its broad spectrum anti-inflammatory activity by a singular mechanism of action, inhibition of protein binding to the cell surface GAG, heparan sulfate. RX-111 and related thieno[2,3-c]pyridine derivatives are potential therapeutics for the treatment of inflammatory and autoimmune diseases.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Cell Line, Tumor
- Colitis/chemically induced
- Colitis/drug therapy
- Colitis/immunology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Heparitin Sulfate/metabolism
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/immunology
- Humans
- Hypersensitivity, Delayed/chemically induced
- Hypersensitivity, Delayed/drug therapy
- Hypersensitivity, Delayed/immunology
- Leukocyte Rolling/drug effects
- Male
- Mice, Inbred BALB C
- Myelin Basic Protein/immunology
- Oxazolone
- Pyridines/pharmacology
- Pyridines/therapeutic use
- Rats
- Rats, Inbred Lew
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Thiophenes/pharmacology
- Thiophenes/therapeutic use
- Treatment Outcome
- Trinitrobenzenesulfonic Acid
Collapse
Affiliation(s)
- Nicholas Harris
- Rimonyx Pharmaceuticals Ltd., Rabin Science Park, 70400, Ness-Ziona, Israel.
- Ephraim Katzir Department of Biotechnology Engineering, ORT Braude Academic College of Engineering, 21982, Karmiel, Israel.
| | - Juraj Koppel
- Institute of Animal Physiology, Slovak Academy of Sciences, 04001, Kosice, Slovakia
| | - Ferenc Zsila
- Biomolecular Self-Assembly Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Stefan Juhas
- Institute of Animal Physiology, Slovak Academy of Sciences, 04001, Kosice, Slovakia
- Institute of Animal Physiology and Genetics of the ASCR, v. v. i., Rumburská 89, 277 21, Liběchov, Czech Republic
| | - Gabriela Il'kova
- Institute of Animal Physiology, Slovak Academy of Sciences, 04001, Kosice, Slovakia
- GYN-FIV a.s., Záhradnícka 42, 821 085, Bratislava, Slovakia
| | | | - Orly Lahmy
- Rimonyx Pharmaceuticals Ltd., Rabin Science Park, 70400, Ness-Ziona, Israel
| | - Gizi Wildbaum
- Department of Immunology, Rappaport Institute, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Nathan Karin
- Department of Immunology, Rappaport Institute, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Regina Zhuk
- Rimonyx Pharmaceuticals Ltd., Rabin Science Park, 70400, Ness-Ziona, Israel
| | - Paul Gregor
- Rimonyx Pharmaceuticals Ltd., Rabin Science Park, 70400, Ness-Ziona, Israel.
- GISMO Therapeutics Inc., A253 ASTECC-UK, Lexington, KY, 40506, USA.
| |
Collapse
|
37
|
Amash A, Wang L, Wang Y, Bhakta V, Fairn GD, Hou M, Peng J, Sheffield WP, Lazarus AH. CD44 Antibody Inhibition of Macrophage Phagocytosis Targets Fcγ Receptor– and Complement Receptor 3–Dependent Mechanisms. THE JOURNAL OF IMMUNOLOGY 2016; 196:3331-40. [DOI: 10.4049/jimmunol.1502198] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/08/2016] [Indexed: 11/19/2022]
|
38
|
Hyaluronan synthesis is necessary for autoreactive T-cell trafficking, activation, and Th1 polarization. Proc Natl Acad Sci U S A 2016; 113:1339-44. [PMID: 26787861 DOI: 10.1073/pnas.1525086113] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The extracellular matrix polysaccharide hyaluronan (HA) accumulates at sites of autoimmune inflammation, including white matter lesions in multiple sclerosis (MS), but its functional importance in pathogenesis is unclear. We have evaluated the impact of 4-methylumbelliferone (4-MU), an oral inhibitor of HA synthesis, on disease progression in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. Treatment with 4-MU decreases the incidence of EAE, delays its onset, and reduces the severity of established disease. 4-MU inhibits the activation of autoreactive T cells and prevents their polarization toward a Th1 phenotype. Instead, 4-MU promotes polarization toward a Th2 phenotpye and induction of Foxp3(+) regulatory T cells. Further, 4-MU hastens trafficking of T cells through secondary lymphoid organs, impairs the infiltration of T cells into the CNS parenchyma, and limits astrogliosis. Together, these data suggest that HA synthesis is necessary for disease progression in EAE and that treatment with 4-MU may be a potential therapeutic strategy in CNS autoimmunity. Considering that 4-MU is already a therapeutic, called hymecromone, that is approved to treat biliary spasm in humans, we propose that it could be repurposed to treat MS.
Collapse
|
39
|
Álvarez-Sánchez N, Cruz-Chamorro I, López-González A, Utrilla JC, Fernández-Santos JM, Martínez-López A, Lardone PJ, Guerrero JM, Carrillo-Vico A. Melatonin controls experimental autoimmune encephalomyelitis by altering the T effector/regulatory balance. Brain Behav Immun 2015; 50:101-114. [PMID: 26130320 DOI: 10.1016/j.bbi.2015.06.021] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/26/2015] [Accepted: 06/26/2015] [Indexed: 02/01/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE), the experimental model for multiple sclerosis (MS), is triggered by myelin-specific Th1 and Th17 cells. The immunomodulatory activities of melatonin have been shown to be beneficial under several conditions in which the immune system is exacerbated. Here, we sought to elucidate the basis of the melatonin protective effect on EAE by characterizing the T effector/regulatory responses, particularly those of the memory cell subsets. Melatonin was tested for its effect on Th1, Th17 and T regulatory (Treg) cells in the lymph nodes and CNS of immunodominant peptide of myelin oligodendrocyte glycoprotein (pMOG)-immunized and EAE mice, respectively. The capacity of melatonin to ameliorate EAE as well as modifying both T cell response and effector/regulatory balance was surveyed. T cell memory subsets and CD44, a key activation marker involved in the EAE pathogenesis, were also examined. Melatonin protected from EAE by decreasing peripheral and central Th1/Th17 responses and enhancing both the Treg frequency and IL-10 synthesis in the CNS. Melatonin reduced the T effector memory population and its pro-inflammatory response and regulated CD44 expression, which was decreased in T effector cells and increased in Tregs. The alterations in the T cell subpopulations were associated with a reduced mononuclear infiltration (CD4 and CD11b cells) of the melatonin-treated mice CNS. For the first time, we report that melatonin protects against EAE by controlling peripheral and central T effector/regulatory responses, effects that might be partially mediated by CD44. This immunomodulatory effect on EAE suggests that melatonin may represent an effective treatment option for MS.
Collapse
Affiliation(s)
- Nuria Álvarez-Sánchez
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Ivan Cruz-Chamorro
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain
| | - Antonio López-González
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Neurosurgery, Virgen Macarena & Virgen del Rocío University Hospitals, Seville, Spain
| | - José C Utrilla
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, Spain
| | - José M Fernández-Santos
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, Spain
| | - Alicia Martínez-López
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain
| | - Patricia J Lardone
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain
| | - Juan M Guerrero
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain; Department of Clinical Biochemistry, Virgen del Rocío University Hospital, Seville, Spain
| | - Antonio Carrillo-Vico
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain.
| |
Collapse
|
40
|
Selectin-mediated leukocyte trafficking during the development of autoimmune disease. Autoimmun Rev 2015; 14:984-95. [DOI: 10.1016/j.autrev.2015.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/18/2015] [Indexed: 12/18/2022]
|
41
|
Weissman I. Evolution of normal and neoplastic tissue stem cells: progress after Robert Hooke. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140364. [PMID: 26416675 PMCID: PMC4633993 DOI: 10.1098/rstb.2014.0364] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2015] [Indexed: 01/29/2023] Open
Abstract
The appearance of stem cells coincides with the transition from single-celled organisms to metazoans. Stem cells are capable of self-renewal as well as differentiation. Each tissue is maintained by self-renewing tissue-specific stem cells. The accumulation of mutations that lead to preleukaemia are in the blood-forming stem cell, while the transition to leukaemia stem cells occurs in the clone at a progenitor stage. All leukaemia and cancer cells escape being removed by scavenger macrophages by expressing the 'don't eat me' signal CD47. Blocking antibodies to CD47 are therapeutics for all cancers, and are currently being tested in clinical trials in the US and UK.
Collapse
Affiliation(s)
- Irving Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
42
|
Imeri F, Blanchard O, Jenni A, Schwalm S, Wünsche C, Zivkovic A, Stark H, Pfeilschifter J, Huwiler A. FTY720 and two novel butterfly derivatives exert a general anti-inflammatory potential by reducing immune cell adhesion to endothelial cells through activation of S1P(3) and phosphoinositide 3-kinase. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:1283-92. [PMID: 26267293 DOI: 10.1007/s00210-015-1159-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 07/27/2015] [Indexed: 10/23/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a key lipid regulator of a variety of cellular responses including cell proliferation and survival, cell migration, and inflammatory reactions. Here, we investigated the effect of S1P receptor activation on immune cell adhesion to endothelial cells under inflammatory conditions. We show that S1P reduces both tumor necrosis factor (TNF)-α- and lipopolysaccharide (LPS)-stimulated adhesion of Jurkat and U937 cells to an endothelial monolayer. The reducing effect of S1P was reversed by the S1P1+3 antagonist VPC23019 but not by the S1P1 antagonist W146. Additionally, knockdown of S1P3, but not S1P1, by short hairpin RNA (shRNA) abolished the reducing effect of S1P, suggesting the involvement of S1P3. A suppression of immune cell adhesion was also seen with the immunomodulatory drug FTY720 and two novel butterfly derivatives ST-968 and ST-1071. On the molecular level, S1P and all FTY720 derivatives reduced the mRNA expression of LPS- and TNF-α-induced adhesion molecules including ICAM-1, VCAM-1, E-selectin, and CD44 which was reversed by the PI3K inhibitor LY294002, but not by the MEK inhibitor U0126.In summary, our data demonstrate a novel molecular mechanism by which S1P, FTY720, and two novel butterfly derivatives acted anti-inflammatory that is by suppressing gene transcription of various endothelial adhesion molecules and thereby preventing adhesion of immune cells to endothelial cells and subsequent extravasation.
Collapse
Affiliation(s)
- Faik Imeri
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Olivier Blanchard
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Aurelio Jenni
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Stephanie Schwalm
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.,Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Christin Wünsche
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.,Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Aleksandra Zivkovic
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine-University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine-University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.
| |
Collapse
|
43
|
McDonald CA, Payne NL, Sun G, Moussa L, Siatskas C, Lim R, Wallace EM, Jenkin G, Bernard CCA. Immunosuppressive potential of human amnion epithelial cells in the treatment of experimental autoimmune encephalomyelitis. J Neuroinflammation 2015; 12:112. [PMID: 26036872 PMCID: PMC4457975 DOI: 10.1186/s12974-015-0322-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/14/2015] [Indexed: 01/25/2023] Open
Abstract
Background Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system (CNS). In recent years, it has been found that cells such as human amnion epithelial cells (hAECs) have the ability to modulate immune responses in vitro and in vivo and can differentiate into multiple cell lineages. Accordingly, we investigated the immunoregulatory effects of hAECs as a potential therapy in an MS-like disease, EAE (experimental autoimmune encephalomyelitis), in mice. Methods Using flow cytometry, the phenotypic profile of hAECs from different donors was assessed. The immunomodulatory properties of hAECs were examined in vitro using antigen-specific and one-way mixed lymphocyte proliferation assays. The therapeutic efficacy of hAECs was examined using a relapsing-remitting model of EAE in NOD/Lt mice. T cell responsiveness, cytokine secretion, T regulatory, and T helper cell phenotype were determined in the peripheral lymphoid organs and CNS of these animals. Results In vitro, hAECs suppressed both specific and non-specific T cell proliferation, decreased pro-inflammatory cytokine production, and inhibited the activation of stimulated T cells. Furthermore, T cells retained their naïve phenotype when co-cultured with hAECs. In vivo studies revealed that hAECs not only suppressed the development of EAE but also prevented disease relapse in these mice. T cell responses and production of the pro-inflammatory cytokine interleukin (IL)-17A were reduced in hAEC-treated mice, and this was coupled with a significant increase in the number of peripheral T regulatory cells and naïve CD4+ T cells. Furthermore, increased proportions of Th2 cells in the peripheral lymphoid organs and within the CNS were observed. Conclusion The therapeutic effect of hAECs is in part mediated by inducing an anti-inflammatory response within the CNS, demonstrating that hAECs hold promise for the treatment of autoimmune diseases like MS.
Collapse
Affiliation(s)
- Courtney A McDonald
- Australian Regenerative Medicine Institute, Monash University, Clayton, 3800, Australia.,The Ritchie Centre, MIMR-PHI Institute of Medical Research, Clayton, VIC, 3800, Australia
| | - Natalie L Payne
- Australian Regenerative Medicine Institute, Monash University, Clayton, 3800, Australia.
| | - Guizhi Sun
- Australian Regenerative Medicine Institute, Monash University, Clayton, 3800, Australia
| | - Leon Moussa
- Australian Regenerative Medicine Institute, Monash University, Clayton, 3800, Australia
| | - Christopher Siatskas
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, VIC, 3800, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Rebecca Lim
- The Ritchie Centre, MIMR-PHI Institute of Medical Research, Clayton, VIC, 3800, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, 3800, Australia
| | - Euan M Wallace
- The Ritchie Centre, MIMR-PHI Institute of Medical Research, Clayton, VIC, 3800, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, 3800, Australia
| | - Graham Jenkin
- The Ritchie Centre, MIMR-PHI Institute of Medical Research, Clayton, VIC, 3800, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, 3800, Australia
| | - Claude C A Bernard
- Australian Regenerative Medicine Institute, Monash University, Clayton, 3800, Australia.
| |
Collapse
|
44
|
Häusler D, Nessler S, Kruse N, Brück W, Metz I. Natalizumab analogon therapy is effective in a B cell-dependent multiple sclerosis model. Neuropathol Appl Neurobiol 2015; 41:814-31. [PMID: 25641089 DOI: 10.1111/nan.12220] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/08/2015] [Indexed: 11/27/2022]
Abstract
AIMS Natalizumab is a humanized monoclonal antibody specific for CD49d receptors of integrins. It inhibits the entry of inflammatory cells into the central nervous system and is approved for the treatment of relapsing-remitting multiple sclerosis (MS). Several lines of evidence indicate an involvement of B cells and plasma cells in MS pathogenesis. However, treatment with the natalizumab analogon PS/2 immunoglobulin G (IgG) has so far only been investigated in T cell-mediated animal models of MS. Due to the importance of B lineage cells in the pathogenesis of MS, the objective of the present study has thus been to analyse the effects of PS/2 IgG in a mouse model of MS with T and B cell cooperation (OSE mice). METHODS OSE mice were treated with the natalizumab analogon PS/2 IgG either at disease onset or after peak of disease. Treatment was also performed with PS/2 F(ab')2 fragments. RESULTS PS/2 IgG treatment improved the clinical outcome and decreased spinal cord demyelination and immune cell infiltration if given early in the disease course. Treatment increased blood leukocytes and resulted in a partial internalization of CD49d in T and B cells. The therapeutic effects of PS/2 IgG injections were independent of the Fc fragment as F(ab')2 injections were equally beneficial. In contrast, PS/2 IgG was not effective when given late in the disease course. CONCLUSIONS Results indicate that natalizumab may also be beneficial in MS with B cell-driven immunopathogenesis.
Collapse
Affiliation(s)
- Darius Häusler
- Department of Neuropathology, University Medical Center, Georg August University, Göttingen, Germany
| | - Stefan Nessler
- Department of Neuropathology, University Medical Center, Georg August University, Göttingen, Germany
| | - Niels Kruse
- Department of Neuropathology, University Medical Center, Georg August University, Göttingen, Germany
| | - Wolfgang Brück
- Department of Neuropathology, University Medical Center, Georg August University, Göttingen, Germany
| | - Imke Metz
- Department of Neuropathology, University Medical Center, Georg August University, Göttingen, Germany
| |
Collapse
|
45
|
Nagy N, Kuipers HF, Frymoyer AR, Ishak HD, Bollyky JB, Wight TN, Bollyky PL. 4-methylumbelliferone treatment and hyaluronan inhibition as a therapeutic strategy in inflammation, autoimmunity, and cancer. Front Immunol 2015; 6:123. [PMID: 25852691 PMCID: PMC4369655 DOI: 10.3389/fimmu.2015.00123] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/06/2015] [Indexed: 12/27/2022] Open
Abstract
Hyaluronan (HA) is a prominent component of the extracellular matrix at many sites of chronic inflammation, including type 1 diabetes (T1D), multiple sclerosis, and numerous malignancies. Recent publications have demonstrated that when HA synthesis is inhibited using 4-methylumbelliferone (4-MU), beneficial effects are observed in several animal models of these diseases. Notably, 4-MU is an already approved drug in Europe and Asia called "hymecromone" where it is used to treat biliary spasm. However, there is uncertainty regarding how 4-MU treatment provides benefit in these animal models and the potential long-term consequences of HA inhibition. Here, we review what is known about how HA contributes to immune dysregulation and tumor progression. Then, we review what is known about 4-MU and hymecromone in terms of mechanism of action, pharmacokinetics, and safety. Finally, we review recent studies detailing the use of 4-MU to treat animal models of cancer and autoimmunity.
Collapse
Affiliation(s)
- Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine , Stanford, CA , USA
| | - Hedwich F Kuipers
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine , Stanford, CA , USA
| | - Adam R Frymoyer
- Department of Pediatrics, Stanford University School of Medicine , Stanford, CA , USA
| | - Heather D Ishak
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine , Stanford, CA , USA
| | - Jennifer B Bollyky
- Department of Pediatrics and Systems Medicine, Stanford University School of Medicine , Stanford, CA , USA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute , Seattle, WA , USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine , Stanford, CA , USA
| |
Collapse
|
46
|
Kodama K, Toda K, Morinaga S, Yamada S, Butte AJ. Anti-CD44 antibody treatment lowers hyperglycemia and improves insulin resistance, adipose inflammation, and hepatic steatosis in diet-induced obese mice. Diabetes 2015; 64:867-75. [PMID: 25294945 PMCID: PMC4392898 DOI: 10.2337/db14-0149] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes (T2D) is a metabolic disease affecting >370 million people worldwide. It is characterized by obesity-induced insulin resistance, and growing evidence has indicated that this causative link between obesity and insulin resistance is associated with visceral adipose tissue inflammation. However, using anti-inflammatory drugs to treat insulin resistance and T2D is not a common practice. We recently applied a bioinformatics methodology to open public data and found that CD44 plays a critical role in the development of adipose tissue inflammation and insulin resistance. In this report, we examined the role of CD44 in T2D by administering daily injections of anti-CD44 monoclonal antibody (mAb) in a high-fat-diet mouse model. Four weeks of therapy with CD44 mAb suppressed visceral adipose tissue inflammation compared with controls and reduced fasting blood glucose levels, weight gain, liver steatosis, and insulin resistance to levels comparable to or better than therapy with the drugs metformin and pioglitazone. These findings suggest that CD44 mAb may be useful as a prototype drug for therapy of T2D by breaking the links between obesity and insulin resistance.
Collapse
Affiliation(s)
- Keiichi Kodama
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA Lucile Packard Children's Hospital, Palo Alto, CA
| | - Kyoko Toda
- Division of Biomedical Research Center, Biomedical Laboratory, Kitasato Institute Hospital, Kitasato University, Minato-ku, Tokyo, Japan
| | - Shojiroh Morinaga
- Department of Diagnostic Pathology, Kitasato Institute Hospital, Kitasato University, Minato-ku, Tokyo, Japan
| | - Satoru Yamada
- Diabetes Center, Kitasato Institute Hospital, Kitasato University, Minato-ku, Tokyo, Japan
| | - Atul J Butte
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA Lucile Packard Children's Hospital, Palo Alto, CA
| |
Collapse
|
47
|
McDonald B, Kubes P. Interactions between CD44 and Hyaluronan in Leukocyte Trafficking. Front Immunol 2015; 6:68. [PMID: 25741341 PMCID: PMC4330908 DOI: 10.3389/fimmu.2015.00068] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/02/2015] [Indexed: 11/17/2022] Open
Abstract
Recruitment of leukocytes from the bloodstream to inflamed tissues requires a carefully regulated cascade of binding interactions between adhesion molecules on leukocytes and endothelial cells. Adhesive interactions between CD44 and hyaluronan (HA) have been implicated in the regulation of immune cell trafficking within various tissues. In this review, the biology of CD44–HA interactions in cell trafficking is summarized, with special attention to neutrophil recruitment within the liver microcirculation. We describe the molecular mechanisms that regulate adhesion between neutrophil CD44 and endothelial HA, including recent evidence implicating serum-derived hyaluronan-associated protein as an important co-factor in the binding of HA to CD44 under flow conditions. CD44–HA-mediated neutrophil recruitment has been shown to contribute to innate immune responses to invading microbes, as well as to the pathogenesis of many inflammatory diseases, including various liver pathologies. As a result, blockade of neutrophil recruitment by targeting CD44–HA interactions has proven beneficial as an anti-inflammatory treatment strategy in a number of animal models of inflammatory diseases.
Collapse
Affiliation(s)
- Braedon McDonald
- Department of Medicine, University of British Columbia , Vancouver, BC , Canada ; Snyder Institute for Chronic Diseases, University of Calgary , Calgary, AB , Canada
| | - Paul Kubes
- Snyder Institute for Chronic Diseases, University of Calgary , Calgary, AB , Canada
| |
Collapse
|
48
|
Chalmin F, Rochemont V, Lippens C, Clottu A, Sailer A, Merkler D, Hugues S, Pot C. Oxysterols regulate encephalitogenic CD4+ T cell trafficking during central nervous system autoimmunity. J Autoimmun 2015; 56:45-55. [DOI: 10.1016/j.jaut.2014.10.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 09/25/2014] [Accepted: 10/08/2014] [Indexed: 12/15/2022]
|
49
|
Mitroulis I, Alexaki VI, Kourtzelis I, Ziogas A, Hajishengallis G, Chavakis T. Leukocyte integrins: role in leukocyte recruitment and as therapeutic targets in inflammatory disease. Pharmacol Ther 2014; 147:123-135. [PMID: 25448040 DOI: 10.1016/j.pharmthera.2014.11.008] [Citation(s) in RCA: 200] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 11/06/2014] [Indexed: 02/06/2023]
Abstract
Infection or sterile inflammation triggers site-specific attraction of leukocytes. Leukocyte recruitment is a process comprising several steps orchestrated by adhesion molecules, chemokines, cytokines and endogenous regulatory molecules. Distinct adhesive interactions between endothelial cells and leukocytes and signaling mechanisms contribute to the temporal and spatial fine-tuning of the leukocyte adhesion cascade. Central players in the leukocyte adhesion cascade include the leukocyte adhesion receptors of the β2-integrin family, such as the αLβ2 and αMβ2 integrins, or of the β1-integrin family, such as the α4β1-integrin. Given the central involvement of leukocyte recruitment in different inflammatory and autoimmune diseases, the leukocyte adhesion cascade in general, and leukocyte integrins in particular, represent key therapeutic targets. In this context, the present review focuses on the role of leukocyte integrins in the leukocyte adhesion cascade. Experimental evidence that has implicated leukocyte integrins as targets in animal models of inflammatory disorders, such as experimental autoimmune encephalomyelitis, psoriasis, inflammatory bone loss and inflammatory bowel disease as well as preclinical and clinical therapeutic applications of antibodies that target leukocyte integrins in various inflammatory disorders are presented. Finally, we review recent findings on endogenous inhibitors that modify leukocyte integrin function, which could emerge as promising therapeutic targets.
Collapse
Affiliation(s)
- Ioannis Mitroulis
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Vasileia I Alexaki
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ioannis Kourtzelis
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Athanassios Ziogas
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - George Hajishengallis
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
50
|
Mueller AM, Yoon BH, Sadiq SA. Inhibition of hyaluronan synthesis protects against central nervous system (CNS) autoimmunity and increases CXCL12 expression in the inflamed CNS. J Biol Chem 2014; 289:22888-22899. [PMID: 24973214 DOI: 10.1074/jbc.m114.559583] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hyaluronan (HA) may have proinflammatory roles in the context of CNS autoimmunity. It accumulates in demyelinated multiple sclerosis (MS) lesions, promotes antigen presentation, and enhances T-cell activation and proliferation. HA facilitates lymphocyte binding to vessels and CNS infiltration at the CNS vascular endothelium. Furthermore, HA signals through Toll-like receptors 2 and 4 to stimulate inflammatory gene expression. We assessed the role of HA in experimental autoimmune encephalomyelitis (EAE), an animal model of MS by administration of 4-methylumbelliferone (4MU), a well established inhibitor of HA synthesis. 4MU decreased hyaluronan synthesis in vitro and in vivo. It was protective in active EAE of C57Bl/6 mice, decreased spinal inflammatory infiltrates and spinal infiltration of Th1 cells, and increased differentiation of regulatory T-cells. In adoptive transfer EAE, feeding of 4MU to donor mice significantly decreased the encephalitogenicity of lymph node cells. The transfer of proteolipid protein (PLP)-stimulated lymph node cells to 4MU-fed mice resulted in a delayed EAE onset and delayed spinal T-cell infiltration. Expression of CXCL12, an anti-inflammatory chemokine, is reduced in MS patients in CSF cells and in spinal cord tissue during EAE. Hyaluronan suppressed production of CXCL12, whereas 4MU increased spinal CXCL12 in naive animals and during neuroinflammation. Neutralization of CXCR4, the most prominent receptor of CXCL12, by administration of AMD3100 diminished the protective impact of 4MU in adoptive transfer EAE. In conclusion, hyaluronan exacerbates CNS autoimmunity, enhances encephalitogenic T-cell responses, and suppresses the protective chemokine CXCL12 in CNS tissue. Inhibition of hyaluronan synthesis with 4MU protects against an animal model of MS and may represent an important therapeutic option in MS and other neuroinflammatory diseases.
Collapse
Affiliation(s)
| | - Bo Hyung Yoon
- Tisch Multiple Sclerosis Research Center of New York, New York, New York 10019
| | - Saud Ahmed Sadiq
- Tisch Multiple Sclerosis Research Center of New York, New York, New York 10019.
| |
Collapse
|