1
|
Vaitaitis GM, Wagner DH. Slow Subcutaneous Release of Glatiramer Acetate or CD40-Targeting Peptide KGYY 6 Is More Advantageous in Treating Ongoing Experimental Autoimmune Encephalomyelitis. Neurol Int 2024; 16:1540-1551. [PMID: 39585073 PMCID: PMC11587089 DOI: 10.3390/neurolint16060114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES One of the first-line disease-modifying treatments of multiple sclerosis (MS) is Glatiramer Acetate (GA), which requires daily or three-times-weekly subcutaneous injections. Disease progression, while slowed, still occurs with time. Increasing the impact of the treatment while decreasing the frequency of injections would be ideal. The mechanism of action of GA remains undefined. We developed an alternate approach, KGYY6, whose mechanism of action targets the CD40 receptor with promising results in an Experimental Autoimmune Encephalomyelitis (EAE) model. METHODS GA and a CD40-targeting peptide, KGYY6, were formulated as slow-release particles used to treat EAE in C57BL/6 mice. RESULTS Compared to liquid formulations, the particle formulations vastly improved drug efficacy in both cases, which would be advantageous in treating MS. GA is a combination of randomly generated peptides, in the size range of 5000-9000 Da, using the amino acids E, A, Y, and K. This approach introduces batch differences that impacts efficacy, a persistent problem with GA. KGYY6 is generated in a controlled process and has a motif, K-YY, which could be generated when manufacturing GA. When testing two different lots of GA or KGYY6, the latter performed equally well across lots, while GA did not. CONCLUSIONS Slow-release formulations of both GA and KGYY6 vastly improve the efficacy of both, and KGYY6 is more consistent in efficacy across different lots.
Collapse
Affiliation(s)
| | - David H. Wagner
- Webb-Waring Center, Department of Medicine, University of Colorado, Anschutz Medical Campus, 12850 E Montview Boulevard, Aurora, CO 80045, USA;
| |
Collapse
|
2
|
Režić Mužinić N, Markotić A, Pavelin S, Polančec D, Buljubašić Šoda M, Bralić A, Šoda J, Mastelić A, Mikac U, Jerković A, Rogić Vidaković M. Expression of CD40 and CD192 in Classical Monocytes in Multiple Sclerosis Patients Assessed with Transcranial Magnetic Stimulation. Biomedicines 2023; 11:2870. [PMID: 37893243 PMCID: PMC10603866 DOI: 10.3390/biomedicines11102870] [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: 09/19/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Expression of CD40 and CD192 markers in different monocyte subpopulations has been reported to be altered in people with MS (pwMS). Also, functional connectivity of the corticospinal motor system pathway alterations has been proved by transcranial magnetic stimulation (TMS). The study objective was to investigate the expression of CD40 and CD192 in classical (CD14++CD16-), intermediate CD14++CD16+ and non-classical (CD14+CD16++) blood monocyte subpopulations in pwMS, undergoing neurophysiological TMS assessment of the corticospinal tract integrity by recording motor-evoked potentials (MEPs). Radiological examination on lesion detection with MRI was performed for 23 patients with relapsing-remitting MS treated with teriflunomide. Then, immunological analysis was conducted on peripheral blood samples collected from the patients and 10 healthy controls (HC). The blood samples were incubated with anti-human CD14, CD16, CD40 and CD192 antibodies. Next, pwMS underwent neurological testing of functional disability (EDSS) and TMS assessment with recording MEPs from upper and lower extremity muscles. The results show that in comparison to HC subjects, both pwMS with normal and altered MEP findings (prolonged MEP latency or absent MEP response) had significantly decreased surface receptor expression measured (MFIs) of CD192 and increased CD40 MFI in classical monocytes, and significantly increased percentages of classical and total monocytes positive for CD40. Knowing CD40's pro-inflammatory action, and CD192 as a molecule that enables the passing of monocytes into the brain, decreased CD192 in classical monocytes could represent a beneficial anti-inflammatory parameter.
Collapse
Affiliation(s)
- Nikolina Režić Mužinić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (A.M.)
| | - Anita Markotić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (A.M.)
| | - Sanda Pavelin
- Department of Neurology, University Hospital of Split, 21000 Split, Croatia
| | | | | | - Antonia Bralić
- Department of Interventional and Diagnostic Radiology, University Hospital of Split, 21000 Split, Croatia
| | - Joško Šoda
- Signal Processing, Analysis, Advanced Diagnostics Research and Education Laboratory (SPAADREL), Department for Marine Electrical Engineering and Information Technologies, Faculty of Maritime Studies, University of Split, 21000 Split, Croatia
| | - Angela Mastelić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (A.M.)
| | - Una Mikac
- Department of Psychology, Faculty of Humanities and Social Sciences, University of Zagreb, 10000 Zagreb, Croatia
| | - Ana Jerković
- Laboratory for Human and Experimental Neurophysiology, Department of Neuroscience, School of Medicine, University of Split, 21000 Split, Croatia
| | - Maja Rogić Vidaković
- Laboratory for Human and Experimental Neurophysiology, Department of Neuroscience, School of Medicine, University of Split, 21000 Split, Croatia
| |
Collapse
|
3
|
Zhu L, Song G, Chen X, Zhang Y, Cui Y, Qiao J, Huang X, Li X, Liu X, Zeng X, Li Y, Wang L, Li B. Higher CD4 +CD40 + T cells (Th40 cells) associate with systemic lupus erythematosus activity. Sci Rep 2023; 13:10702. [PMID: 37400575 DOI: 10.1038/s41598-023-37749-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023] Open
Abstract
The aim of this study was to investigate the characteristics of CD4+CD40+ T cells (Th40 cells) in Chinese systemic lupus erythematosus (SLE) patients. Flow cytometry was used to identify the percentage of Th40 cells in peripheral blood from 24 SLE patients and 24 healthy individuals and the level of IL-2, IL-4, IL-6, IL-10, IFN-r, and TNF-α in serum (22 cases) from the SLE patients. Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2000) was used to assess the SLE disease active state. The percentage of Th40 cells in T cells from SLE patients (19.37 ± 17.43) (%) was significantly higher than that from healthy individuals (4.52 ± 3.16) (%) (P < 0.001). The percentage of Th40 cells was also positively associated with SLEDAI-2000 (P = 0.001) and negatively associated with complement C3 (P = 0.007). The Th40 cell percentage was different in SLE patients with different organs involved. The Th40 cell percentage in SLE patients with lupus serositis (29.29 ± 22.19) was significantly higher than that in patients without serositis (13.41 ± 10.79; P = 0.040), and the percentage in SLE patients with lupus pneumonia involvement (29.11 ± 11.88) was significantly higher than that in patients without lupus pneumonia (16.80 ± 17.99; P = 0.043). After 4 weeks treatment, the Th40 cell percentage decreased significantly (P = 0.005). However, Th40 cell expression was not related to cytokines (IL-2, IL-4, IL-6, IL-10, IFN-r, and TNF-α; P > 0.05). A significantly higher percentage of Th40 cells was found in SLE patients, and the Th40 cell percentage was associated with SLE activity. Thus, Th40 cells may be used as a predictor for SLE disease activity and severity and therapeutic efficacy.
Collapse
Affiliation(s)
- Lihua Zhu
- Department of Rheumatology and Immunology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Guangmei Song
- Department of Rheumatology and Immunology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Xiaohui Chen
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yue Zhang
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yanjie Cui
- Department of Rheumatology and Immunology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Jie Qiao
- Department of Rheumatology and Immunology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Xinran Huang
- Department of Rheumatology and Immunology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Xueqin Li
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiaoen Liu
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiangbo Zeng
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yangqiu Li
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Liang Wang
- Department of Oncology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
| | - Bo Li
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
4
|
Vaitaitis GM, Wagner DH. Modulating CD40 and integrin signaling in the proinflammatory nexus using a 15-amino-acid peptide, KGYY 15. J Biol Chem 2023; 299:104625. [PMID: 36944397 PMCID: PMC10141526 DOI: 10.1016/j.jbc.2023.104625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
CD40 signaling has long been a target in autoimmunity. Attempts to block signaling between CD40 and CD154 during clinical trials using monoclonal antibodies suffered severe adverse events. Previously, we developed a peptide, KGYY15, that targets CD40 and, in preclinical trials, prevents type 1 diabetes in >90% of cases and reverses new-onset hyperglycemia in 56% of cases. It did so by establishing normal effector T-cell levels rather than ablating the cells and causing immunosuppression. However, the relationship between KGYY15 and other elements of the complex signaling network of CD40 is not clear. Studying interactions between proteins from autoimmune and nonautoimmune mice, we demonstrate interactions between CD40 and integrin CD11a/CD18, which complicates the understanding of the inflammatory nexus and how to prevent autoinflammation. In addition to interacting with CD40, KGYY15 interacts with the integrins CD11a/CD18 and CD11b/CD18. We argue that modulation of CD40-CD154 signaling may be more advantageous than complete inhibition because it may preserve normal immunity to pathogens.
Collapse
Affiliation(s)
- Gisela M Vaitaitis
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David H Wagner
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
5
|
Curran C, Vaitaitis G, Waid D, Volmer T, Alverez E, Wagner DH. Ocrevus reduces TH40 cells, a biomarker of systemic inflammation, in relapsing multiple sclerosis (RMS) and in progressive multiple sclerosis (PMS). J Neuroimmunol 2023; 374:578008. [PMID: 36535240 PMCID: PMC9868100 DOI: 10.1016/j.jneuroim.2022.578008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/16/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Treating MS has been difficult. One successful drug is Ocrelizumab (anti-CD20), used for the chronic relapsing MS (RMS) and the progressive MS (PMS) forms. TH40 cells are pathogenic effector T cells that increase in percentage and numbers during chronic inflammation. Here we show that in the earliest MS course, clinically isolated syndrome (CIS), TH40 cells expand in number. In PMS TH40 cell numbers remain expanded demonstrating sustained chronic inflammation. In RMS TH40 cells were found in CSF and express CD20. Ocrelizumab reduced TH40 cells to healthy control levels in patients. During treatment inflammatory cytokine producing TH40 cells were decreased.
Collapse
Affiliation(s)
- Christian Curran
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Gisela Vaitaitis
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Dan Waid
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Timothy Volmer
- The Department of Neurology, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Enrique Alverez
- The Department of Neurology, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - David H Wagner
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America.
| |
Collapse
|
6
|
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
|
7
|
Lei TY, Ye YZ, Zhu XQ, Smerin D, Gu LJ, Xiong XX, Zhang HF, Jian ZH. The immune response of T cells and therapeutic targets related to regulating the levels of T helper cells after ischaemic stroke. J Neuroinflammation 2021; 18:25. [PMID: 33461586 PMCID: PMC7814595 DOI: 10.1186/s12974-020-02057-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/09/2020] [Indexed: 12/21/2022] Open
Abstract
Through considerable effort in research and clinical studies, the immune system has been identified as a participant in the onset and progression of brain injury after ischaemic stroke. Due to the involvement of all types of immune cells, the roles of the immune system in stroke pathology and associated effects are complicated. Past research concentrated on the functions of monocytes and neutrophils in the pathogenesis of ischaemic stroke and tried to demonstrate the mechanisms of tissue injury and protection involving these immune cells. Within the past several years, an increasing number of studies have elucidated the vital functions of T cells in the innate and adaptive immune responses in both the acute and chronic phases of ischaemic stroke. Recently, the phenotypes of T cells with proinflammatory or anti-inflammatory function have been demonstrated in detail. T cells with distinctive phenotypes can also influence cerebral inflammation through various pathways, such as regulating the immune response, interacting with brain-resident immune cells and modulating neurogenesis and angiogenesis during different phases following stroke. In view of the limited treatment options available following stroke other than tissue plasminogen activator therapy, understanding the function of immune responses, especially T cell responses, in the post-stroke recovery period can provide a new therapeutic direction. Here, we discuss the different functions and temporal evolution of T cells with different phenotypes during the acute and chronic phases of ischaemic stroke. We suggest that modulating the balance between the proinflammatory and anti-inflammatory functions of T cells with distinct phenotypes may become a potential therapeutic approach that reduces the mortality and improves the functional outcomes and prognosis of patients suffering from ischaemic stroke.
Collapse
Affiliation(s)
- Tian-Yu Lei
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Ying-Ze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Xi-Qun Zhu
- Department of Head and Neck and Neurosurgery, Hubei Cancer Hospital, Wuhan, 430079, Hubei Province, People's Republic of China
| | - Daniel Smerin
- University of Central Florida College of Medicine, Orlando, FL, 32827, USA
| | - Li-Juan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Xiao-Xing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Zhi-Hong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
8
|
Knoop J, Eugster A, Gavrisan A, Lickert R, Sedlmeier EM, Dietz S, Lindner A, Warncke K, Hummel N, Ziegler AG, Bonifacio E. Maternal Type 1 Diabetes Reduces Autoantigen-Responsive CD4 + T Cells in Offspring. Diabetes 2020; 69:661-669. [PMID: 31896551 DOI: 10.2337/db19-0751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/28/2019] [Indexed: 11/13/2022]
Abstract
Autoimmunity against pancreatic β-cell autoantigens is a characteristic of childhood type 1 diabetes (T1D). Autoimmunity usually appears in genetically susceptible children with the development of autoantibodies against (pro)insulin in early childhood. The offspring of mothers with T1D are protected from this process. The aim of this study was to determine whether the protection conferred by maternal T1D is associated with improved neonatal tolerance against (pro)insulin. Consistent with improved neonatal tolerance, the offspring of mothers with T1D had reduced cord blood CD4+ T-cell responses to proinsulin and insulin, a reduction in the inflammatory profile of their proinsulin-responsive CD4+ T cells, and improved regulation of CD4+ T cell responses to proinsulin at 9 months of age, as compared with offspring with a father or sibling with T1D. Maternal T1D was also associated with a modest reduction in CpG methylation of the INS gene in cord blood mononuclear cells from offspring with a susceptible INS genotype. Our findings support the concept that a maternal T1D environment improves neonatal immune tolerance against the autoantigen (pro)insulin.
Collapse
Affiliation(s)
- Jan Knoop
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anne Eugster
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Anita Gavrisan
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Ramona Lickert
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Eva-Maria Sedlmeier
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Sevina Dietz
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Annett Lindner
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Katharina Warncke
- Department of Pediatrics, Klinikum Rechts der Isar, School of Medicine, Technical University Munich, Munich, Germany
| | - Nadine Hummel
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
- Forschergruppe Diabetes e.V., Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| |
Collapse
|
9
|
Vaitaitis GM, Yussman MG, Wagner DH. A CD40 targeting peptide prevents severe symptoms in experimental autoimmune encephalomyelitis. J Neuroimmunol 2019; 332:8-15. [PMID: 30925295 PMCID: PMC6535109 DOI: 10.1016/j.jneuroim.2019.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 01/07/2023]
Abstract
CD40/CD154-interaction is critical in the development of Experimental Autoimmune Encephalomyelitis (EAE; mouse model of Multiple Sclerosis). Culprit CD4+CD40+ T cells drive a more severe form of EAE than conventional CD4 T cells. Blocking CD40/CD154-interaction with CD154-antibody prevents or ameliorates disease but had thrombotic complications in clinical trials. We targeted CD40 using a CD154-sequence based peptide. Peptides in human therapeutics demonstrate good safety. A small peptide, KGYY6, ameliorates EAE when given as pretreatment or at first symptoms. KGYY6 binds Th40 and memory T cells, affecting expression of CD69 and IL-10 in the CD4 T cell compartment, ultimately hampering disease development.
Collapse
Affiliation(s)
- Gisela M Vaitaitis
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Martin G Yussman
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - David H Wagner
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
10
|
Aarts SABM, Seijkens TTP, Kusters PJH, van Tiel CM, Reiche ME, den Toom M, Beckers L, van Roomen CPAA, de Winther MPJ, Kooij G, Lutgens E. Macrophage CD40 signaling drives experimental autoimmune encephalomyelitis. J Pathol 2019; 247:471-480. [PMID: 30471110 PMCID: PMC6519352 DOI: 10.1002/path.5205] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/23/2018] [Accepted: 11/19/2018] [Indexed: 01/06/2023]
Abstract
The costimulatory CD40L-CD40 dyad plays a major role in multiple sclerosis (MS). CD40 is highly expressed on MHCII+ B cells, dendritic cells and macrophages in human MS lesions. Here we investigated the role of the CD40 downstream signaling intermediates TNF receptor-associated factor 2 (TRAF2) and TRAF6 in MHCII+ cells in experimental autoimmune encephalomyelitis (EAE). Both MHCII-CD40-Traf2-/- and MHCII-CD40-Traf6-/- mice showed a reduction in clinical signs of EAE and prevented demyelination. However, only MHCII-CD40-Traf6-/- mice displayed a decrease in myeloid and lymphoid cell infiltration into the CNS that was accompanied by reduced levels of TNF-α, IL-6 and IFN-γ. As CD40-TRAF6 interactions predominantly occur in macrophages, we subjected CD40flfl LysMcre mice to EAE. This myeloid-specific deletion of CD40 resulted in a significant reduction in EAE severity, reduced CNS inflammation and demyelination. In conclusion, the CD40-TRAF6 signaling pathway in MHCII+ cells plays a key role in neuroinflammation and demyelination during EAE. Concomitant with the fact that CD40-TRAF6 interactions are predominant in macrophages, depletion of myeloid CD40 also reduces neuroinflammation. CD40-TRAF6 interactions thus represent a promising therapeutic target for MS. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Suzanne ABM Aarts
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Tom TP Seijkens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU)MunichGermany
| | - Pascal JH Kusters
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Claudia M van Tiel
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Myrthe E Reiche
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Cindy PAA van Roomen
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Menno PJ de Winther
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU)MunichGermany
- Department of Molecular Cell Biology and ImmunologyAmsterdam University Medical Centers, MS Center Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and ImmunologyAmsterdam University Medical Centers, MS Center Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU)MunichGermany
| |
Collapse
|
11
|
Vaitaitis GM, Wagner DH. Are we aiming to miss in translational autoimmunity treatments? F1000Res 2019; 7:1754. [PMID: 30542620 PMCID: PMC6259590 DOI: 10.12688/f1000research.16894.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/28/2018] [Indexed: 11/20/2022] Open
Abstract
Autoimmunity treatments, fruitfully pioneered in mouse models, can be disappointing or result in immunosuppression and opportunistic infections in translational trials. Many possible reasons exist, but one major, overlooked reason may be the treatment timing in relation to circadian oscillations of the immune system. Mice and humans both have immunological circadian clocks and experience the same circulatory oscillations of immune cells with regards to their sleep/wake phases, but have opposite sleep/wake phases with regard to the daylight cycle. Therefore, researchers mainly study mice and potential autoimmunity treatments during the murine sleep/rest phase, which is when pro-inflammatory mediators and more adaptive immune cells are prevalent in the circulation. In translational trials, however, treatment administration happens primarily during a patient's wake/activity phase, during the daytime, which is when more local and acute immune responses are active in the circulation. Therefore, we believe that the most opportune window for autoimmunity treatment may be missed in translational trials. Shifting the timing, and adjusting dosing to target only immune cells that are active at that time, may result in higher success with minimized immunosuppression or toxicities.
Collapse
Affiliation(s)
- Gisela M Vaitaitis
- Webb-Waring Center, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.,Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - David H Wagner
- Webb-Waring Center, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.,Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
12
|
Aarts SABM, Seijkens TTP, van Dorst KJF, Dijkstra CD, Kooij G, Lutgens E. The CD40-CD40L Dyad in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. Front Immunol 2017; 8:1791. [PMID: 29312317 PMCID: PMC5732943 DOI: 10.3389/fimmu.2017.01791] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/29/2017] [Indexed: 12/16/2022] Open
Abstract
The CD40-CD40L dyad is an immune checkpoint regulator that promotes both innate and adaptive immune responses and has therefore an essential role in the development of inflammatory diseases, including multiple sclerosis (MS). In MS, CD40 and CD40L are expressed on immune cells present in blood and lymphoid organs, affected resident central nervous system (CNS) cells, and inflammatory cells that have infiltrated the CNS. CD40-CD40L interactions fuel the inflammatory response underlying MS, and both genetic deficiency and antibody-mediated inhibition of the CD40-CD40L dyad reduce disease severity in experimental autoimmune encephalomyelitis (EAE). Both proteins are therefore attractive therapeutic candidates to modulate aberrant inflammatory responses in MS. Here, we discuss the genetic, experimental and clinical studies on the role of CD40 and CD40L interactions in EAE and MS and we explore novel approaches to therapeutically target this dyad to combat neuroinflammatory diseases.
Collapse
Affiliation(s)
- Suzanne A. B. M. Aarts
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Tom T. P. Seijkens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany
| | | | - Christine D. Dijkstra
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany
| |
Collapse
|
13
|
Vaitaitis GM, Waid DM, Yussman MG, Wagner DH. CD40-mediated signalling influences trafficking, T-cell receptor expression, and T-cell pathogenesis, in the NOD model of type 1 diabetes. Immunology 2017; 152:243-254. [PMID: 28542921 DOI: 10.1111/imm.12761] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/27/2017] [Accepted: 05/08/2017] [Indexed: 12/23/2022] Open
Abstract
CD40 plays a critical role in the pathogenesis of type 1 diabetes (T1D). The mechanism of action, however, is undetermined, probably because CD40 expression has been grossly underestimated. CD40 is expressed on numerous cell types that now include T cells and pancreatic β cells. CD40+ CD4+ cells [T helper type 40 (TH40)] prove highly pathogenic in NOD mice and in translational human T1D studies. We generated BDC2.5.CD40-/- and re-derived NOD.CD154-/- mice to better understand the CD40 mechanism of action. Fully functional CD40 expression is required not only for T1D development but also for insulitis. In NOD mice, TH40 cell expansion in pancreatic lymph nodes occurs before insulitis and demonstrates an activated phenotype compared with conventional CD4+ cells, apparently regardless of antigen specificity. TH40 T-cell receptor (TCR) usage demonstrates increases in several Vα and Vβ species, particularly Vα3.2+ that arise early and are sustained throughout disease development. TH40 cells isolated from diabetic pancreas demonstrate a relatively broad TCR repertoire rather than restricted clonal expansions. The expansion of the Vα/Vβ species associated with diabetes depends upon CD40 signalling; NOD.CD154-/- mice do not expand the same TCR species. Finally, CD40-mediated signals significantly increase pro-inflammatory Th1- and Th17-associated cytokines whereas CD28 co-stimulus alternatively promotes regulatory cytokines.
Collapse
Affiliation(s)
- Gisela M Vaitaitis
- The Webb-Waring Center, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dan M Waid
- The Webb-Waring Center, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Martin G Yussman
- The Webb-Waring Center, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David H Wagner
- The Webb-Waring Center, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Division of Pulmonary Sciences and Critical Care, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
14
|
Matrine promotes oligodendrocyte development in CNS autoimmunity through the PI3K/Akt signaling pathway. Life Sci 2017; 180:36-41. [PMID: 28499934 DOI: 10.1016/j.lfs.2017.05.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/03/2017] [Accepted: 05/09/2017] [Indexed: 01/21/2023]
Abstract
AIMS Matrine (MAT), a quinolizidine alkaloid derived from the herb Radix Sophorae flavescens, has been recently found to be beneficial in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, mainly through its anti-inflammatory effect. In the present study, we tested the effect of MAT on ongoing EAE and defined possible mechanisms underlying its effects on myelination and oligodendrocytes. MAIN METHODS EAE was induced in C57BL/6 mice and MAT treatment was started at disease onset. Clinical scores were monitored daily; spinal cords and the corpus callosum brain region of mice were harvested on day 23 p.i. for inflammatory infiltration and demyelination of the central nervous system. Myelin content and the development of oligodendrocytes and their precursors were determined by immunostaining, and expression of p-Akt, p-mTOR, p-PI3K, and p-P70S6 was determined by Western blot. KEY FINDINGS MAT effectively suppressed EAE severity and increased the expression of proteolipid protein, a myelin protein that is a marker of CNS myelin. MAT treatment largely increased the number of mature oligodendrocytes, and significantly activated the PI3K/Akt/mTOR signaling pathway, which is required for oligodendrocyte survival and axon myelination. SIGNIFICANCE These findings demonstrate a beneficial effect of MAT on oligodendrocyte differentiation and myelination during EAE, most likely through activating the PI3K/Akt/mTOR signaling pathway.
Collapse
|
15
|
Mouat IC, Morse ZJ, Jean-Baptiste VSE, Allanach JR, Horwitz MS. Fresh Ideas, Foundational Experiments (FIFE): Immunology and Diabetes 2016 FIFE Symposium. Front Endocrinol (Lausanne) 2017; 8:238. [PMID: 28974943 PMCID: PMC5610696 DOI: 10.3389/fendo.2017.00238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/30/2017] [Indexed: 12/16/2022] Open
Abstract
The first Fresh Ideas, Foundational Experiments (FIFE): Immunology and Diabetes symposia workshop took place in 2016 and exemplified the active interest of a number of several investigators interested the global rise in the incidence of type 1 diabetes (T1D). This increase does not correlate with genetic drift and indicates that environmental exposures are playing an increasingly significant role. Despite major biomedical and technological advances in diagnosis and treatment, treatments are frequently insufficient as they do not inhibit the progression of the underlying autoimmune response and often fail to prevent life-threatening complications. T1D is the result of autoimmune destruction of the insulin-producing beta cells of the pancreas, and the precise, mechanistic contribution of the immune system to disease pathogenesis and progression remains to be fully characterized. Ultimately, the combinatorial effect of concurrent factors, including beta cell fragility, exogenous stressors, and genetic priming of the innate and adaptive immune system, work together to induce T1D autoimmunity. Thus, T1D is the result of immunological defects and environmental pathogens, requiring the sustained attention of collaborative research teams such as FIFE: I & D with varied perspectives, unified by the universally held goal of finding a sustainable, life-long cure. Herein, the authors provide perspective on various fields in T1D research highlighted by speakers participating in the inaugural FIFE symposium.
Collapse
Affiliation(s)
- Isobel C. Mouat
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Zachary J. Morse
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | | | - Jessica R. Allanach
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Marc S. Horwitz
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Marc S. Horwitz,
| |
Collapse
|
16
|
Wagner DH. Overlooked Mechanisms in Type 1 Diabetes Etiology: How Unique Costimulatory Molecules Contribute to Diabetogenesis. Front Endocrinol (Lausanne) 2017; 8:208. [PMID: 28878738 PMCID: PMC5572340 DOI: 10.3389/fendo.2017.00208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/08/2017] [Indexed: 01/16/2023] Open
Abstract
Type 1 Diabetes (T1D) develops when immune cells invade the pancreatic islets resulting in loss of insulin production in beta cells. T cells have been proven to be central players in that process. What is surprising, however, is that classic mechanisms of tolerance cannot explain diabetogenesis; alternate mechanisms must now be considered. T cell receptor (TCR) revision is the process whereby T cells in the periphery alter TCR expression, outside the safety-net of thymic selection pressures. This process results in an expanded T cell repertoire, capable of responding to a universe of pathogens, but limitations are that increased risk for autoimmune disease development occurs. Classic T cell costimulators including the CD28 family have long been thought to be the major drivers for full T cell activation. In actuality, CD28 and its family member counterparts, ICOS and CTLA-4, all drive regulatory responses. Inflammation is driven by CD40, not CD28. CD40 as a costimulus has been largely overlooked. When naïve T cells interact with antigen presenting cell CD154, the major ligand for CD40, is induced. This creates a milieu for T cell (CD40)-T cell (CD154) interaction, leading to inflammation. Finally, defined pathogenic effector cells including TH40 (CD4+CD40+) cells can express FOXP3 but are not Tregs. The cells loose FOXP3 to become pathogenic effector cells. Each of these mechanisms creates novel options to better understand diabetogenesis and create new therapeutic targets for T1D.
Collapse
Affiliation(s)
- David H. Wagner
- The Program in Integrated Immunology, Department of Medicine, Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: David H. Wagner Jr.,
| |
Collapse
|