51
|
Heming M, Schulte-Mecklenbeck A, Brix T, Wolbert J, Ruland T, Klotz L, Meuth SG, Gross CC, Wiendl H, Meyer Zu Hörste G. Immune Cell Profiling of the Cerebrospinal Fluid Provides Pathogenetic Insights Into Inflammatory Neuropathies. Front Immunol 2019; 10:515. [PMID: 30984164 PMCID: PMC6448021 DOI: 10.3389/fimmu.2019.00515] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/26/2019] [Indexed: 01/17/2023] Open
Abstract
Objective: Utilize immune cell profiles in the cerebrospinal fluid (CSF) to advance the understanding and potentially support the diagnosis of inflammatory neuropathies. Methods: We analyzed CSF cell flow cytometry data of patients with definite Guillain-Barré syndrome (GBS, n = 26) and chronic inflammatory demyelinating polyneuropathy (CIDP, n = 32) based on established diagnostic criteria in comparison to controls with relapsing-remitting multiple sclerosis (RRMS, n = 49) and idiopathic intracranial hypertension (IIH, n = 63). Results: Flow cytometry revealed disease-specific changes of CSF cell composition with a significant increase of NKT cells and CD8+ T cells in CIDP, NK cells in GBS, and B cells and plasma cells in MS in comparison to IIH controls. Principal component analysis demonstrated distinct CSF immune cells pattern in inflammatory neuropathies vs. RRMS. Systematic receiver operator curve (ROC) analysis identified NKT cells as the best parameter to distinguish GBS from CIDP. Composite scores combing several of the CSF parameters differentiated inflammatory neuropathies from IIH and GBS from CIDP with high confidence. Applying a novel dimension reduction technique, we observed an intra-disease heterogeneity of inflammatory neuropathies. Conclusion: Inflammatory neuropathies display disease- and subtype-specific alterations of CSF cell composition. The increase of NKT cells and CD8+ T cells in CIDP and NK cells in GBS, suggests a central role of cytotoxic cell types in inflammatory neuropathies varying between acute and chronic subtypes. Composite scores constructed from multi-dimensional CSF parameters establish potential novel diagnostic tools. Intra-disease heterogeneity suggests distinct disease mechanisms in subgroups of inflammatory neuropathies.
Collapse
Affiliation(s)
- Michael Heming
- Department of Neurology, Institute of Translational Neurology, University of Münster, Münster, Germany
| | | | - Tobias Brix
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Jolien Wolbert
- Department of Neurology, Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Tillmann Ruland
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Luisa Klotz
- Department of Neurology, Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Catharina C Gross
- Department of Neurology, Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Gerd Meyer Zu Hörste
- Department of Neurology, Institute of Translational Neurology, University of Münster, Münster, Germany
| |
Collapse
|
52
|
ADAM10 in Alzheimer's disease: Pharmacological modulation by natural compounds and its role as a peripheral marker. Biomed Pharmacother 2019; 113:108661. [PMID: 30836275 DOI: 10.1016/j.biopha.2019.108661] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) represents a global burden in the economics of healthcare systems. Amyloid-β (Aβ) peptides are formed by amyloid-β precursor protein (AβPP) cleavage, which can be processed by two pathways. The cleavage by the α-secretase A Disintegrin And Metalloprotease 10 (ADAM10) releases the soluble portion (sAβPPα) and prevents senile plaques. This pathway remains largely unknown and ignored, mainly regarding pharmacological approaches that may act via different signaling cascades and thus stimulate non-amyloidogenic cleavage through ADAM10. This review emphasizes the effects of natural compounds on ADAM10 modulation, which eventuates in a neuroprotective mechanism. Moreover, ADAM10 as an AD biomarker is revised. New treatments and preventive interventions targeting ADAM10 regulation for AD are necessary, considering the wide variety of ADAM10 substrates.
Collapse
|
53
|
Khoenkhoen S, Erikson E, Ádori M, Stark JM, Scholz JL, Cancro MP, Pedersen GK, Karlsson Hedestam GB. TACI expression and plasma cell differentiation are impaired in the absence of functional IκBNS. Immunol Cell Biol 2019; 97:485-497. [PMID: 30597621 PMCID: PMC6850186 DOI: 10.1111/imcb.12228] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/17/2018] [Accepted: 12/27/2018] [Indexed: 12/15/2022]
Abstract
Impaired classical NF‐κB pathway signaling causes reduced antibody responses to T‐independent (TI) antigens. We investigated the potential reasons for defective TI responses in mice lacking the atypical inhibitory kappa B (IκB) protein of the NF‐κB pathway, IκBNS. Analyses of the plasma cell compartment in vitro and in vivo after challenge with lipopolysaccharide (LPS) showed significant decreases in the frequencies of plasma cells in the absence of IκBNS. In vitro activation of B cells via the B cell receptor or via Toll‐like receptor 4 revealed that early activation events were unaffected in IκBNS‐deficient B cells, while proliferation was reduced compared to in similarly stimulated wildtype (wt) B cells. IκBNS‐deficient B cells also displayed impaired upregulation of the transmembrane activator and calcium modulator cyclophilin ligand interactor (TACI), which is essential for TI responses, and decreased sensitivity to TACI ligands upon stimulation. Furthermore, IκBNS‐deficient B cells, in contrast to wt B cells, displayed altered expression of IRF4, Blimp‐1 and Pax5 upon LPS‐induced differentiation, indicating impaired transcriptional regulation of plasma cell generation.
Collapse
Affiliation(s)
- Sharesta Khoenkhoen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Elina Erikson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Monika Ádori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Julian M Stark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jean L Scholz
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Cancro
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriel K Pedersen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
54
|
Lendak DF, Mihajlović DM, Novakov-Mikić AS, Boban JM, Ubavić M, Brkić SV. APRIL and sTACI could be predictors of multiorgan dysfunction syndrome in sepsis. Virulence 2018; 9:946-953. [PMID: 29781374 PMCID: PMC7000195 DOI: 10.1080/21505594.2018.1462636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 03/30/2018] [Accepted: 04/05/2018] [Indexed: 12/29/2022] Open
Abstract
Although the role of B cells in sepsis immunoregulation has become an interesting topic, there is lack of data on the role of B cell function regulators in prediction of multiorgan dysfunction syndrome (MODS). The aim of this study was to evaluate the prognostic value of A Proliferation Inducing Ligand (APRIL) and soluble Transmembrane Activator and CAML Interactor Protein (sTACI), the main B cell function regulators, in prediction of MODS development within the first 48 h after admission to intensive care unit, among septic patients. We included 112 patients with sepsis, treated at Clinic for Infectious Diseases and Emergency Center, Clinical Center of Vojvodina, Novi Sad, Serbia. Plasma concentrations of APRIL and sTACI were determined at the admission and potential development of MODS was confirmed in the first 48 h. Concentrations of APRIL (p = 0.003) and sTACI (p<0.001) were higher in patients who developed MODS (n = 30). ROC curve analysis showed that AUC for sTACI (AUC = 0.764) was greater than that for procalcitonin (AUC = 0.719) and APRIL (AUC = 0.673) in MODS development prediction. Multivariate regression analysis showed that sTACI, as an anti-inflammatory biomarker stimulating the apoptosis of B cells, was the only independent predictor of MODS, beside SOFA score. Elevated level of sTACI could be the alarm for the increased B cell apoptosis and development of immune paralysis. Including these biomarkers into predictive scores specific for septic patients may potentially improve their sensitivity and specificity. Measurement of their concentrations dynamics could contribute to better assessment of sepsis evolution and timely introduction of immunomodulatory therapy.
Collapse
Affiliation(s)
- Dajana F. Lendak
- Department of Infectious Diseases, Faculty of Medicine, University of Novi Sad; Clinic for Infectious Diseases, Clinical centre of Vojvodina; Hajduk Veljkova 3, Novi Sad, Serbia
| | - Dunja M. Mihajlović
- Department of Anesthesiology and Perioperative Medicine, Faculty of Medicine, University of Novi Sad; Emergency Center, Clinical centre of Vojvodina, Hajduk Veljkova 3, Novi Sad, Serbia
| | - Aleksandra S. Novakov-Mikić
- Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Novi Sad; Clinical Center of Vojvodina, Clinic of Gynecology and Obstetrics, Hajduk Veljkova 1, Novi Sad, Serbia
| | - Jasmina M. Boban
- Department of Radiology, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 1, Novi Sad, Serbia
| | - Milan Ubavić
- Health Care Institution for Laboratory Diagnostics „Medlab“, Ilije Ognjanovića 1, Novi Sad, Serbia
| | - Snežana V. Brkić
- Department of Infectious Diseases, Faculty of Medicine, University of Novi Sad; Clinic for Infectious Diseases, Clinical centre of Vojvodina; Hajduk Veljkova 3, Novi Sad, Serbia
- Department of Infectious Diseases, Faculty of Medicine, University of Novi Sad, Clinic for Infectious Diseases, Clinical centre of Vojvodina, Hajduk Veljkova 1, Novi Sad, Serbia
| |
Collapse
|
55
|
Smulski CR, Eibel H. BAFF and BAFF-Receptor in B Cell Selection and Survival. Front Immunol 2018; 9:2285. [PMID: 30349534 PMCID: PMC6186824 DOI: 10.3389/fimmu.2018.02285] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/14/2018] [Indexed: 12/16/2022] Open
Abstract
The BAFF-receptor (BAFFR) is encoded by the TNFRSF13C gene and is one of the main pro-survival receptors in B cells. Its function is impressively documented in humans by a homozygous deletion within exon 2, which leads to an almost complete block of B cell development at the stage of immature/transitional B cells. The resulting immunodeficiency is characterized by B-lymphopenia, agammaglobulinemia, and impaired humoral immune responses. However, different from mutations affecting pathway components coupled to B cell antigen receptor (BCR) signaling, BAFFR-deficient B cells can still develop into IgA-secreting plasma cells. Therefore, BAFFR deficiency in humans is characterized by very few circulating B cells, very low IgM and IgG serum concentrations but normal or high IgA levels.
Collapse
Affiliation(s)
- Cristian R Smulski
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Freiburg, Germany
| | - Hermann Eibel
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
56
|
Hohlfeld R, Meinl E. Ocrelizumab in multiple sclerosis: markers and mechanisms. Lancet Neurol 2018; 16:259-261. [PMID: 28327329 DOI: 10.1016/s1474-4422(17)30048-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 11/18/2022]
Affiliation(s)
- Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospitals and Biomedical Center, Ludwig Maximilians University, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospitals and Biomedical Center, Ludwig Maximilians University, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
57
|
Oost W, Talma N, Meilof JF, Laman JD. Targeting senescence to delay progression of multiple sclerosis. J Mol Med (Berl) 2018; 96:1153-1166. [PMID: 30229272 PMCID: PMC6208951 DOI: 10.1007/s00109-018-1686-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/18/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is a chronic and often progressive, demyelinating disease of the central nervous system (CNS) white and gray matter and the single most common cause of disability in young adults. Age is one of the factors most strongly influencing the course of progression in MS. One of the hallmarks of aging is cellular senescence. The elimination of senescent cells with senolytics has very recently been shown to delay age-related dysfunction in animal models for other neurological diseases. In this review, the possible link between cellular senescence and the progression of MS is discussed, and the potential use of senolytics as a treatment for progressive MS is explored. Currently, there is no cure for MS and there are limited treatment options to slow the progression of MS. Current treatment is based on immunomodulatory approaches. Various cell types present in the CNS can become senescent and thus potentially contribute to MS disease progression. We propose that, after cellular senescence has indeed been shown to be directly implicated in disease progression, administration of senolytics should be tested as a potential therapeutic approach for the treatment of progressive MS.
Collapse
Affiliation(s)
- Wendy Oost
- University of Groningen, Groningen, The Netherlands
| | - Nynke Talma
- European Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan F Meilof
- Department of Neurology, Martini Hospital, Groningen, The Netherlands.,MS Center Noord Nederland (MSCNN), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands. .,MS Center Noord Nederland (MSCNN), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
58
|
Meinl E, Thaler FS, Lichtenthaler SF. Shedding of BAFF/APRIL Receptors Controls B Cells. Trends Immunol 2018; 39:673-676. [DOI: 10.1016/j.it.2018.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/28/2018] [Accepted: 07/06/2018] [Indexed: 01/10/2023]
|
59
|
Lendak DF, Mihajlović DM, Novakov-Mikić AS, Mitić IM, Boban JM, Brkić SV. The role of TNF-α superfamily members in immunopathogenesis of sepsis. Cytokine 2018; 111:125-130. [PMID: 30142533 DOI: 10.1016/j.cyto.2018.08.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/11/2018] [Accepted: 08/13/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Members of TNFα superfamily, A proliferation inducing ligand (APRIL), B-cell activating factor (BAFF) and Transmembrane activator and calcium cyclophylin interactor (TACI) are main regulators of B-cell function. The aim of this study was to evaluate concentrations of APRIL, BAFF and soluble TACI (sTACI) receptor in septic patients compared to healthy controls and compare concentrations of these biomarkers depending on sepsis severity and outcome. MATERIALS AND METHODS A total of 115 septic patients and 30 healthy volunteers were included and concentrations of APRIL, BAFF and sTACI were determined in all subjects at the admission (ELISA R&D Systems tests). Concentrations of these biomarkers in function of sepsis severity (sepsis n = 94 and septic shock n = 21) and outcome (lethal n = 40, recovery n = 75) were tested, as well as correlations with APACHE II and SOFA scores, immunoglobulins, complement, PCT and CRP concentrations. RESULTS Concentrations of all three biomarkers were significantly increased in septic patients compared to controls (AUCAPRIL = 0.982, AUCBAFF = 0.873, AUCsTACI = 0.683). Higher concentrations of APRIL and sTACI (p = 0.033, p = 0.037), and lower concentrations of BAFF (p = 0.005) were observed in patients with septic shock compared to sepsis. BAFF concentrations correlated positively with IgM, C3 and C4 levels. sTACI and APRIL were shown to be predictors of lethal outcome (p = 0.003, p = 0.049). CONCLUSIONS Concentrations of observedTNFα superfamily members are significantly increased in septic patients, confirming their role in sepsis pathogenesis.Higher concentrations of anti-inflammatory sTACI receptor correlated with severity of sepsis and poorer prognosis, thus potentially indicating domination of anti-inflammatory response in septic patients with worse outcome.
Collapse
Affiliation(s)
- Dajana F Lendak
- Department of Infectious Diseases, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia; Clinical Centre of Vojvodina, Novi Sad, Serbia.
| | - Dunja M Mihajlović
- Clinical Centre of Vojvodina, Novi Sad, Serbia; Department of Anestesiology and Perioerative Medicine, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Aleksandra S Novakov-Mikić
- Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia; Department of Internal Medicine (Immunology), Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Igor M Mitić
- Clinical Centre of Vojvodina, Novi Sad, Serbia; Department of Internal Medicine (Immunology), Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Jasmina M Boban
- Department for Radiology, Faculty of Medicine, University of Novi Sad, Vojvodina, Serbia; Center for Imaging Diagnostics, Institute of Oncology, Sremska Kamenica, Serbia
| | - Snežana V Brkić
- Department of Infectious Diseases, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia; Clinical Centre of Vojvodina, Novi Sad, Serbia
| |
Collapse
|
60
|
Thaler FS, Laurent SA, Huber M, Mulazzani M, Dreyling M, Ködel U, Kümpfel T, Straube A, Meinl E, von Baumgarten L. Soluble TACI and soluble BCMA as biomarkers in primary central nervous system lymphoma. Neuro Oncol 2018; 19:1618-1627. [PMID: 28521029 DOI: 10.1093/neuonc/nox097] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background B-cell survival is regulated through interactions of B-cell-activating factor and a proliferation-inducing ligand with their receptors transmembrane activator and CAML interactor (TACI) and B-cell maturation antigen (BCMA). We evaluated the diagnostic potential of soluble TACI (sTACI) and soluble BCMA (sBCMA) in CSF and serum as biomarkers in primary CNS lymphoma (PCNSL). Methods CSF (n = 176) and serum samples (n = 105) from patients with clinically or radiologically suspected PCNSL as well as from control patients were collected prospectively. Levels of sTACI and sBCMA were analyzed by enzyme-linked immunosorbent assay. Additionally, in patients with PCNSL, CSF was analyzed during disease course (time of diagnosis, n = 26; relapse, n = 10; remission, n = 14), and in 2 patients long-term longitudinal analysis was performed. Results Soluble TACI and sBCMA are significantly increased in patients with PCNSL (sTACI, median: 445 pg/mL; sBCMA, median: 760 pg/mL) compared with control patients (sTACI, median: 0 pg/mL; sBCMA, median: 290 pg/mL). At a cutoff value of 68.4 pg/mL, sTACI shows high sensitivity (87.9%) and specificity (88.3%) for the diagnosis of active PCNSL. Soluble BCMA is less sensitive (72.7%) and specific (71.8%) (cutoff: 460 pg/mL). When both markers are combined, specificity increases, however, at the cost of a lower sensitivity. In serum, both sTACI and sBCMA are not increased in PCNSL patients. Both soluble receptors correlate with clinical course and therapy response. Conclusions Our results suggest that sTACI and sBCMA in the CSF are promising new biomarkers for diagnosis and therapy monitoring in PCNSL. However, our findings need to be validated in an independent cohort.
Collapse
Affiliation(s)
- Franziska S Thaler
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| | - Sarah A Laurent
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| | - Marion Huber
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| | - Matthias Mulazzani
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| | - Martin Dreyling
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| | - Uwe Ködel
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| | - Andreas Straube
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| | - Louisa von Baumgarten
- Institute of Clinical Neuroimmunology, Biomedical Center and Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Neurology, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany; Department of Medicine III, Klinikum Grosshadern, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
61
|
Zou F, Wang X, Han X, Rothschild G, Zheng SG, Basu U, Sun J. Expression and Function of Tetraspanins and Their Interacting Partners in B Cells. Front Immunol 2018; 9:1606. [PMID: 30072987 PMCID: PMC6058033 DOI: 10.3389/fimmu.2018.01606] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/27/2018] [Indexed: 01/26/2023] Open
Abstract
Tetraspanins are transmembrane proteins that modulate multiple diverse biological processes, including signal transduction, cell–cell communication, immunoregulation, tumorigenesis, cell adhesion, migration, and growth and differentiation. Here, we provide a systematic review of the involvement of tetraspanins and their partners in the regulation and function of B cells, including mechanisms associated with antigen presentation, antibody production, cytokine secretion, co-stimulator expression, and immunosuppression. Finally, we direct our focus to the signaling mechanisms, evolutionary conservation aspects, expression, and potential therapeutic strategies that could be based on tetraspanins and their interacting partners.
Collapse
Affiliation(s)
- Fagui Zou
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xu Wang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xinxin Han
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Gerson Rothschild
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Song Guo Zheng
- Department of Medicine, Milton S. Hershey Medical Center at Penn State University, Pennsylvania, PA, United States.,Center for Clinic Immunology, Third Affiliated Hospital at Sun Yat-Sen University, Guangzhou, China
| | - Uttiya Basu
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Jianbo Sun
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
62
|
Matthews AL, Koo CZ, Szyroka J, Harrison N, Kanhere A, Tomlinson MG. Regulation of Leukocytes by TspanC8 Tetraspanins and the "Molecular Scissor" ADAM10. Front Immunol 2018; 9:1451. [PMID: 30013551 PMCID: PMC6036176 DOI: 10.3389/fimmu.2018.01451] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/12/2018] [Indexed: 01/16/2023] Open
Abstract
A disintegrin and metalloproteinase 10 (ADAM10) is a ubiquitous transmembrane protein that functions as a "molecular scissor" to cleave the extracellular regions from its transmembrane target proteins. ADAM10 is well characterized as the ligand-dependent activator of Notch proteins, which control cell fate decisions. Indeed, conditional knockouts of ADAM10 in mice reveal impaired B-, T-, and myeloid cell development and/or function. ADAM10 cleaves many other leukocyte-expressed substrates. On B-cells, ADAM10 cleavage of the low-affinity IgE receptor CD23 promotes allergy and asthma, cleavage of ICOS ligand impairs antibody responses, and cleavage of the BAFF-APRIL receptor transmembrane activator and CAML interactor, and BAFF receptor, reduce B-cell survival. On microglia, increased ADAM10 cleavage of a rare variant of the scavenger receptor triggering receptor expressed on myeloid cells 2 may increase susceptibility to Alzheimer's disease. We and others recently showed that ADAM10 interacts with one of six different regulatory tetraspanin membrane proteins, which we termed the TspanC8 subgroup, comprising Tspan5, Tspan10, Tspan14, Tspan15, Tspan17, and Tspan33. The TspanC8s are required for ADAM10 exit from the endoplasmic reticulum, and emerging evidence suggests that they dictate ADAM10 subcellular localization and substrate specificity. Therefore, we propose that ADAM10 should not be regarded as a single scissor, but as six different scissors with distinct substrate specificities, depending on the associated TspanC8. In this review, we collate recent transcriptomic data to present the TspanC8 repertoires of leukocytes, and we discuss the potential role of the six TspanC8/ADAM10 scissors in leukocyte development and function.
Collapse
Affiliation(s)
- Alexandra L Matthews
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Chek Ziu Koo
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Justyna Szyroka
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Neale Harrison
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Aditi Kanhere
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Michael G Tomlinson
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
63
|
The role of the BAFF/APRIL system in the T cell-independent specific response to blood stage Plasmodium falciparum hemozoin. Cytokine 2018; 111:445-453. [PMID: 29884307 DOI: 10.1016/j.cyto.2018.05.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND The B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) are tumor necrosis factor family members that regulate B cell maturation, proliferation, survival and function. We have previously shown that blood-stage Plasmodium falciparum hemozoin (HZ) can act as a T-independent antigen (TI Ag) that induces the production of specific IgG to soluble crude P. falciparum Ag through the BAFF pathway. However, we have not yet clarified whether HZ need APRIL signaling in the TI response. Here, we aimed to clarify whether both BAFF and APRIL signaling pathways play roles in HZ induction of specific antibody production without T-cell help. METHODS Normal monocytes alone or co-cultured with naïve B cells were stimulated by HZ (10 µM) in vitro. Naïve B cell cultures, with HZ alone or with exogenous recombinant BAFF (rBAFF) and recombinant APRIL (rAPRIL) plus recombinant IL-4 (rIL-4) for 6 and 10 days were used as controls to investigate activation of B cells. At various times, the levels of sBAFF, sAPRIL, and HZ-specific IgG in the culture supernatants were assessed by enzyme-linked immunosorbent assay. The BAFF and APRIL expression levels on the HZ-stimulated monocytes and their specific receptors on activated B cells, including the BAFF receptor (BAFF-R), the transmembrane activator and calcium-modulator and cyclophilin ligand interactor (TACI) and the B cell maturation antigen (BCMA), were determined by flow cytometry. mRNA expression levels for the receptors were validated using Real-Time quantitative PCR. RESULTS HZ-activated monocytes released sBAFF and sAPRIL during the 72 h stimulation period. Increased mRNA encoding of their cognate receptors, BAFF-R, TACI, and BCMA, and increased HZ-specific IgG levels were also observed in HZ induction within the monocyte and B cell co-culture. The experiments under control conditions revealed that HZ alone could induce B cell culture to produce a small amount of the specific IgG compared with those in medium alone or rBAFF + rAPRIL + rIL-4. CONCLUSION Taken together, we suggest that in the TI response HZ stimulates monocyte and B cell co-culture to produce specific IgG through BAFF, APRIL and other independent complimentary signaling pathways.
Collapse
|
64
|
Brummer T, Pigoni M, Rossello A, Wang H, Noy PJ, Tomlinson MG, Blobel CP, Lichtenthaler SF. The metalloprotease ADAM10 (a disintegrin and metalloprotease 10) undergoes rapid, postlysis autocatalytic degradation. FASEB J 2018; 32:3560-3573. [PMID: 29430990 DOI: 10.1096/fj.201700823rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The transmembrane protein, ADAM10 (a disintegrin and metalloprotease 10), has key physiologic functions-for example, during embryonic development and in the brain. During transit through the secretory pathway, immature ADAM10 (proADAM10) is converted into its proteolytically active, mature form (mADAM10). Increasing or decreasing the abundance and/or activity of mADAM10 is considered to be a therapeutic approach for the treatment of such diseases as Alzheimer's disease and cancer. Yet biochemical detection and characterization of mADAM10 has been difficult. In contrast, proADAM10 is readily detected-for example, in immunoblots-which suggests that mADAM10 is only a fraction of total cellular ADAM10. Here, we demonstrate that mADAM10, but not proADAM10, unexpectedly undergoes rapid, time-dependent degradation upon biochemical cell lysis in different cell lines and in primary neurons, which prevents the detection of the majority of mADAM10 in immunoblots. This degradation required the catalytic activity of ADAM10, was efficiently prevented by adding active site inhibitors to the lysis buffer, and did not affect proADAM10, which suggests that ADAM10 degradation occurred in an intramolecular and autoproteolytic manner. Inhibition of postlysis autoproteolysis demonstrated efficient cellular ADAM10 maturation with higher levels of mADAM10 than proADAM10. Moreover, a cycloheximide chase experiment revealed that mADAM10 is a long-lived protein with a half-life of approximately 12 h. In summary, our study demonstrates that mADAM10 autoproteolysis must be blocked to allow for the proper detection of mADAM10, which is essential for the correct interpretation of biochemical and cellular studies of ADAM10.-Brummer, T., Pigoni, M., Rossello, A., Wang, H., Noy, P. J., Tomlinson, M. G., Blobel, C. P., Lichtenthaler, S. F. The metalloprotease ADAM10 (a disintegrin and metalloprotease 10) undergoes rapid, postlysis autocatalytic degradation.
Collapse
Affiliation(s)
- Tobias Brummer
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Martina Pigoni
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Huanhuan Wang
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Peter J Noy
- School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Carl P Blobel
- Hospital for Special Surgery, Research Institute, New York, New York, USA.,Department of Medicine, Weill Cornell Medicine, New York, New York, USA.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York, USA.,Institute for Advanced Study, Technische Universität München, Munich, Germany
| | - Stefan F Lichtenthaler
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
65
|
Bao X, Shi J, Xie F, Liu Z, Yu J, Chen W, Zhang Z, Xu Q. Proteolytic Release of the p75NTR Intracellular Domain by ADAM10 Promotes Metastasis and Resistance to Anoikis. Cancer Res 2018; 78:2262-2276. [DOI: 10.1158/0008-5472.can-17-2789] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/22/2017] [Accepted: 01/30/2018] [Indexed: 11/16/2022]
|
66
|
Miyazaki Y, Niino M, Takahashi E, Suzuki M, Mizuno M, Hisahara S, Fukazawa T, Amino I, Nakano F, Nakamura M, Akimoto S, Minami N, Fujiki N, Doi S, Shimohama S, Terayama Y, Kikuchi S. Fingolimod induces BAFF and expands circulating transitional B cells without activating memory B cells and plasma cells in multiple sclerosis. Clin Immunol 2018; 187:95-101. [DOI: 10.1016/j.clim.2017.10.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/15/2017] [Accepted: 10/23/2017] [Indexed: 01/01/2023]
|
67
|
Samy E, Wax S, Huard B, Hess H, Schneider P. Targeting BAFF and APRIL in systemic lupus erythematosus and other antibody-associated diseases. Int Rev Immunol 2017; 36:3-19. [PMID: 28215100 DOI: 10.1080/08830185.2016.1276903] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The B cell-stimulating molecules, BAFF (B cell activating factor) and APRIL (a proliferation-inducing ligand), are critical factors in the maintenance of the B cell pool and humoral immunity. In addition, BAFF and APRIL are involved in the pathogenesis of a number of human autoimmune diseases, with elevated levels of these cytokines detected in the sera of patients with systemic lupus erythematosus (SLE), IgA nephropathy, Sjögren's syndrome, and rheumatoid arthritis. As such, both molecules are rational targets for new therapies in B cell-driven autoimmune diseases, and several inhibitors of BAFF or BAFF and APRIL together have been investigated in clinical trials. These include the BAFF/APRIL dual inhibitor, atacicept, and the BAFF inhibitor, belimumab, which is approved as an add-on therapy for patients with active SLE. Post hoc analyses of these trials indicate that baseline serum levels of BAFF and BAFF/APRIL correlate with treatment response to belimumab and atacicept, respectively, suggesting a role for the two molecules as predictive biomarkers. It will, however, be important to refine future testing to identify active forms of BAFF and APRIL in the circulation, as well as to distinguish between homotrimer and heteromer configurations. In this review, we discuss the rationale for dual BAFF/APRIL inhibition versus single BAFF inhibition in autoimmune disease, by focusing on the similarities and differences between the physiological and pathogenic roles of the two molecules. A summary of the preclinical and clinical data currently available is also presented.
Collapse
Affiliation(s)
- Eileen Samy
- a EMD Serono Research & Development Institute, Inc. , Billerica , Massachusetts , USA
| | - Stephen Wax
- a EMD Serono Research & Development Institute, Inc. , Billerica , Massachusetts , USA
| | - Bertrand Huard
- b Institute for Advanced Biosciences , University Grenoble Alpes , INSERM U1209, Grenoble , France
| | | | - Pascal Schneider
- d Department of Biochemistry , University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
68
|
Borhis G, Trovato M, Chaoul N, Ibrahim HM, Richard Y. B-Cell-Activating Factor and the B-Cell Compartment in HIV/SIV Infection. Front Immunol 2017; 8:1338. [PMID: 29163465 PMCID: PMC5663724 DOI: 10.3389/fimmu.2017.01338] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/03/2017] [Indexed: 12/12/2022] Open
Abstract
With the goal to design effective HIV vaccines, intensive studies focused on broadly neutralizing antibodies, which arise in a fraction of HIV-infected people. Apart from identifying new vulnerability sites in the viral envelope proteins, these studies have shown that a fraction of these antibodies are produced by self/poly-reactive B-cells. These findings prompted us to revisit the B-cell differentiation and selection process during HIV/SIV infection and to consider B-cells as active players possibly shaping the helper T-cell program within germinal centers (GCs). In this context, we paid a particular attention to B-cell-activating factor (BAFF), a key cytokine in B-cell development and immune response that is overproduced during HIV/SIV infection. As it does in autoimmune diseases, BAFF excess might contribute to the abnormal rescue of self-reactive B-cells at several checkpoints of the B-cell development and impair memory B-cell generation and functions. In this review, we first point out what is known about the functions of BAFF/a proliferation-inducing ligand and their receptors [B-cell maturation, transmembrane activator and CAML interactor (TACI), and BAFF-R], in physiological and pathophysiological settings, in mice and humans. In particular, we highlight recent results on the previously underappreciated regulatory functions of TACI and on the highly regulated production of soluble TACI and BAFF-R that act as decoy receptors. In light of recent data on BAFF, TACI, and BAFF-R, we then revisit the altered phenotypes and functions of B-cell subsets during the acute and chronic phase of HIV/SIV infection. Given the atypical phenotype and reduced functions of memory B-cells in HIV/SIV infection, we particularly discuss the GC reaction, a key checkpoint where self-reactive B-cells are eliminated and pathogen-specific memory B-cells and plasmablasts/cells are generated in physiological settings. Through its capacity to differentially bind and process BAFF-R and TACI on GC B-cells and possibly on follicular helper T-cells, BAFF appears as a key regulator of the physiological GC reaction. Its local excess during HIV/SIV infection could play a key role in B-cell dysregulations.
Collapse
Affiliation(s)
- Gwenoline Borhis
- INSERM u1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris-Descartes, Paris, France
| | - Maria Trovato
- INSERM u1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris-Descartes, Paris, France
| | - Nada Chaoul
- Commissariat à l’Energie Atomique, Institut des maladies Emergentes et Thérapies innovantes, Service d’Immuno-Virologie, Fontenay-aux Roses, France
| | - Hany M. Ibrahim
- INSERM u1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris-Descartes, Paris, France
| | - Yolande Richard
- INSERM u1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris-Descartes, Paris, France
| |
Collapse
|
69
|
Abstract
The two ligands B cell-activating factor of the tumor necrosis factor family (BAFF) and a proliferation-inducing ligand (APRIL) and the three receptors BAFF receptor (BAFF-R), transmembrane activator and calcium-modulating cyclophilin ligand interactor (TACI), and B cell maturation antigen (BCMA) are members of the "BAFF system molecules." BAFF system molecules are primarily involved in B cell homeostasis. The relevance of BAFF system molecules in host responses to microbial assaults has been investigated in clinical studies and in mice deficient for each of these molecules. Many microbial products modulate the expression of these molecules. Data from clinical studies suggest a correlation between increased expression levels of BAFF system molecules and elevated B cell responses. Depending on the pathogen, heightened B cell responses may strengthen the host response or promote susceptibility. Whereas pathogen-mediated increases in the expression levels of the ligands and/or the receptors appear to promote microbial clearance, certain pathogens have evolved to ablate B cell responses by suppressing the expression of TACI and/or BAFF-R on B cells. Other than its well-established role in B cell responses, the TACI-mediated activation of macrophages is also implicated in resistance to intracellular pathogens. An improved understanding of the role that BAFF system molecules play in infection may assist in devising novel strategies for vaccine development.
Collapse
Affiliation(s)
- Jiro Sakai
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mustafa Akkoyunlu
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
70
|
Circulating B cells in type 1 diabetics exhibit fewer maturation-associated phenotypes. Clin Immunol 2017; 183:336-343. [PMID: 28951327 DOI: 10.1016/j.clim.2017.09.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/16/2017] [Accepted: 09/22/2017] [Indexed: 01/05/2023]
Abstract
Although autoantibodies have been used for decades as diagnostic and prognostic markers in type 1 diabetes (T1D), further analysis of developmental abnormalities in B cells could reveal tolerance checkpoint defects that could improve individualized therapy. To evaluate B cell developmental progression in T1D, immunophenotyping was used to classify circulating B cells into transitional, mature naïve, mature activated, and resting memory subsets. Then each subset was analyzed for the expression of additional maturation-associated markers. While the frequencies of B cell subsets did not differ significantly between patients and controls, some T1D subjects exhibited reduced proportions of B cells that expressed transmembrane activator and CAML interactor (TACI) and Fas receptor (FasR). Furthermore, some T1D subjects had B cell subsets with lower frequencies of class switching. These results suggest circulating B cells exhibit variable maturation phenotypes in T1D. These phenotypic variations may correlate with differences in B cell selection in individual T1D patients.
Collapse
|
71
|
Friebus-Kardash J, Branco L, Ribi C, Chizzolini C, Huynh-Do U, Dubler D, Roux-Lombard P, Dolff S, Kribben A, Eisenberger U, Trendelenburg M. Immune complexes containing serum B-cell activating factor and immunoglobulin G correlate with disease activity in systemic lupus erythematosus. Nephrol Dial Transplant 2017; 33:54-64. [DOI: 10.1093/ndt/gfx220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/22/2017] [Indexed: 12/31/2022] Open
|
72
|
Pérez-Martínez CA, Maravillas-Montero JL, Meza-Herrera I, Vences-Catalán F, Zlotnik A, Santos-Argumedo L. Tspan33 is Expressed in Transitional and Memory B Cells, but is not Responsible for High ADAM10 Expression. Scand J Immunol 2017; 86:23-30. [PMID: 28449222 DOI: 10.1111/sji.12559] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/18/2017] [Indexed: 12/27/2022]
Abstract
Tetraspanins are a family of transmembrane proteins that form membrane microdomains. They play important roles in migration, adhesion and other cellular processes. TspanC8, a subfamily of tetraspanins, was found to associate and promote ADAM10 trafficking and cell surface localization. One of its members, Tspan33, is expressed in activated B cells. Using RT-PCR and flow cytometry, we analysed the pattern of expression of Tspan33 in B cells from healthy donors. We found Tspan33 expression in early and late stages of B cell development. However, Tspan33 expression did not correlate with ADAM10 surface expression. We also found expression of Tspan33 early in the activation process. Given its predominant expression in activated B cells and in several lymphomas, but not in naive B cells, we hypothesize that Tspan33 could be a potential target for therapeutic purposes.
Collapse
Affiliation(s)
- C A Pérez-Martínez
- Departamento de Biomedicina Molecular, CINVESTAV-IPN, CDMX, Mexico City, Mexico.,Facultad de Medicina, UNAM, CDMX, Mexico City, Mexico
| | | | - I Meza-Herrera
- Departamento de Biomedicina Molecular, CINVESTAV-IPN, CDMX, Mexico City, Mexico
| | - F Vences-Catalán
- Departamento de Biomedicina Molecular, CINVESTAV-IPN, CDMX, Mexico City, Mexico.,Department of Medicine, Division of Oncology, Stanford University Medical Center, Stanford, CA, USA
| | - A Zlotnik
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - L Santos-Argumedo
- Departamento de Biomedicina Molecular, CINVESTAV-IPN, CDMX, Mexico City, Mexico
| |
Collapse
|
73
|
Wetzel S, Seipold L, Saftig P. The metalloproteinase ADAM10: A useful therapeutic target? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [PMID: 28624438 DOI: 10.1016/j.bbamcr.2017.06.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteolytic cleavage represents a unique and irreversible posttranslational event regulating the function and half-life of many intracellular and extracellular proteins. The metalloproteinase ADAM10 has raised attention since it cleaves an increasing number of protein substrates close to the extracellular membrane leaflet. This "ectodomain shedding" regulates the turnover of a number of transmembrane proteins involved in cell adhesion and receptor signaling. It can initiate intramembrane proteolysis followed by nuclear transport and signaling of the cytoplasmic domain. ADAM10 has also been implicated in human disorders ranging from neurodegeneration to dysfunction of the immune system and cancer. Targeting proteases for therapeutic purposes remains a challenge since these enzymes including ADAM10 have a wide range of substrates. Accelerating or inhibiting a specific protease activity is in most cases associated with unwanted side effects and a therapeutic useful window of application has to be carefully defined. A better understanding of the regulatory mechanisms controlling the expression, subcellular localization and activity of ADAM10 will likely uncover suitable drug targets which will allow a more specific and fine-tuned modulation of its proteolytic activity.
Collapse
Affiliation(s)
- Sebastian Wetzel
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Lisa Seipold
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany.
| |
Collapse
|
74
|
Endres K, Deller T. Regulation of Alpha-Secretase ADAM10 In vitro and In vivo: Genetic, Epigenetic, and Protein-Based Mechanisms. Front Mol Neurosci 2017; 10:56. [PMID: 28367112 PMCID: PMC5355436 DOI: 10.3389/fnmol.2017.00056] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/20/2017] [Indexed: 12/21/2022] Open
Abstract
ADAM10 (A Disintegrin and Metalloproteinase 10) has been identified as the major physiological alpha-secretase in neurons, responsible for cleaving APP in a non-amyloidogenic manner. This cleavage results in the production of a neuroprotective APP-derived fragment, APPs-alpha, and an attenuated production of neurotoxic A-beta peptides. An increase in ADAM10 activity shifts the balance of APP processing toward APPs-alpha and protects the brain from amyloid deposition and disease. Thus, increasing ADAM10 activity has been proposed an attractive target for the treatment of neurodegenerative diseases and it appears to be timely to investigate the physiological mechanisms regulating ADAM10 expression. Therefore, in this article, we will (1) review reports on the physiological regulation of ADAM10 at the transcriptional level, by epigenetic factors, miRNAs and/or protein interactions, (2) describe conditions, which change ADAM10 expression in vitro and in vivo, (3) report how neuronal ADAM10 expression may be regulated in humans, and (4) discuss how this knowledge on the physiological and pathophysiological regulation of ADAM10 may help to preserve or restore brain function.
Collapse
Affiliation(s)
- Kristina Endres
- Clinic of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg-University Mainz Mainz, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt/Main, Germany
| |
Collapse
|
75
|
Tien WS, Chen JH, Wu KP. SheddomeDB: the ectodomain shedding database for membrane-bound shed markers. BMC Bioinformatics 2017; 18:42. [PMID: 28361715 PMCID: PMC5374707 DOI: 10.1186/s12859-017-1465-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A number of membrane-anchored proteins are known to be released from cell surface via ectodomain shedding. The cleavage and release of membrane proteins has been shown to modulate various cellular processes and disease pathologies. Numerous studies revealed that cell membrane molecules of diverse functional groups are subjected to proteolytic cleavage, and the released soluble form of proteins may modulate various signaling processes. Therefore, in addition to the secreted protein markers that undergo secretion through the secretory pathway, the shed membrane proteins may comprise an additional resource of noninvasive and accessible biomarkers. In this context, identifying the membrane-bound proteins that will be shed has become important in the discovery of clinically noninvasive biomarkers. Nevertheless, a data repository for biological and clinical researchers to review the shedding information, which is experimentally validated, for membrane-bound protein shed markers is still lacking. RESULTS In this study, the database SheddomeDB was developed to integrate publicly available data of the shed membrane proteins. A comprehensive literature survey was performed to collect the membrane proteins that were verified to be cleaved or released in the supernatant by immunological-based validation experiments. From 436 studies on shedding, 401 validated shed membrane proteins were included, among which 199 shed membrane proteins have not been annotated or validated yet by existing cleavage databases. SheddomeDB attempted to provide a comprehensive shedding report, including the regulation of shedding machinery and the related function or diseases involved in the shedding events. In addition, our published tool ShedP was embedded into SheddomeDB to support researchers for predicting the shedding event on unknown or unrecorded membrane proteins. CONCLUSIONS To the best of our knowledge, SheddomeDB is the first database for the identification of experimentally validated shed membrane proteins and currently may provide the most number of membrane proteins for reviewing the shedding information. The database included membrane-bound shed markers associated with numerous cellular processes and diseases, and some of these markers are potential novel markers because they are not annotated or validated yet in other databases. SheddomeDB may provide a useful resource for discovering membrane-bound shed markers. The interactive web of SheddomeDB is publicly available at http://bal.ym.edu.tw/SheddomeDB/ .
Collapse
Affiliation(s)
- Wei-Sheng Tien
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.,Bioinformatics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan
| | - Jun-Hong Chen
- Department of Computer Science, National Taipei University of Education, Taipei, 106, Taiwan
| | - Kun-Pin Wu
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
76
|
Schuh E, Musumeci A, Thaler FS, Laurent S, Ellwart JW, Hohlfeld R, Krug A, Meinl E. Human Plasmacytoid Dendritic Cells Display and Shed B Cell Maturation Antigen upon TLR Engagement. THE JOURNAL OF IMMUNOLOGY 2017; 198:3081-3088. [DOI: 10.4049/jimmunol.1601746] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/13/2017] [Indexed: 01/06/2023]
|
77
|
Smulski CR, Kury P, Seidel LM, Staiger HS, Edinger AK, Willen L, Seidl M, Hess H, Salzer U, Rolink AG, Rizzi M, Schneider P, Eibel H. BAFF- and TACI-Dependent Processing of BAFFR by ADAM Proteases Regulates the Survival of B Cells. Cell Rep 2017; 18:2189-2202. [DOI: 10.1016/j.celrep.2017.02.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/23/2016] [Accepted: 01/30/2017] [Indexed: 11/26/2022] Open
|
78
|
|
79
|
Interferon-β therapy specifically reduces pathogenic memory B cells in multiple sclerosis patients by inducing a FAS-mediated apoptosis. Immunol Cell Biol 2016; 94:886-894. [PMID: 27265253 DOI: 10.1038/icb.2016.55] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/06/2016] [Accepted: 05/29/2016] [Indexed: 12/27/2022]
Abstract
Growing evidences put B lymphocytes on a central stage in multiple sclerosis (MS) immunopathology. While investigating the effects of interferon-β (IFN-β) therapy, one of the most used first-line disease-modifying drugs for the treatment of relapsing-remitting MS, in circulating B-cell sub-populations, we found a specific and marked decrease of CD27+ memory B cells. Interestingly, memory B cells are considered a population with a great disease-driving relevance in MS and resulted to be also target of B-cell depleting therapies. In addition, Epstein-Barr virus (EBV), associated with MS etiopathogenesis, harbors in this cell type and an IFN-β-induced reduction of the memory B-cell compartment, in turn, resulted in a decreased expression of the EBV gene latent membrane protein 2A in treated patients. We found that in vivo IFN-β therapy specifically and highly induced apoptosis in memory B cells, in accordance with a strong increase of the apoptotic markers Annexin-V and active caspase-3, via a mechanism requiring the FAS-receptor/TACI (transmembrane activator and CAML interactor) signaling. Thus, efficacy of IFN-β therapy in MS may rely not only on its recognized anti-inflammatory activities but also on the specific depletion of memory B cells, considered to be a pathogenic cell subset, reducing their inflammatory impact in target organs.
Collapse
|
80
|
Iyer AS, Khaskhely NM, Leggat DJ, Ohtola JA, Saul-McBeth JL, Khuder SA, Westerink MAJ. Inflammatory Markers and Immune Response to Pneumococcal Vaccination in HIV-Positive and -Negative Adults. PLoS One 2016; 11:e0150261. [PMID: 26930208 PMCID: PMC4773189 DOI: 10.1371/journal.pone.0150261] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/01/2016] [Indexed: 11/30/2022] Open
Abstract
Background Members of the Tumor Necrosis Factor (TNF)-superfamily have speculated roles in the response against T-independent type II antigens (TI-II) including pneumococcal polysaccharides (PPS). Dysregulation in their expression is associated with an enhanced risk for pneumococcal disease in neonates but their expression in other high-risk populations including HIV-positive individuals remains to be elucidated. Objective To investigate signals that contribute towards PPS-response and identify potential anomalies that may account for diminished serological response in HIV-positive individuals post Pneumovax (PPV23) immunization. Methods Markers of inflammation, C-reactive protein (CRP), IL-6, sCD27 and sCD30, were assessed in HIV-positive and -negative individuals as potential predictors of PPV23 response. Serum levels of B cell activating factor (BAFF), transmembrane activator and calcium-modulator and cytophilin ligand interactor (TACI), B cell maturation antigen (BCMA) and B cell expression of BAFF-R, TACI, BCMA, CD40 and CD21 were assessed in total (unselected) and PPS23F (antigen)-specific B cells of PPV23 immunized HIV-positive and -negative individuals. Results CRP, sCD27, sCD30 and BAFF were significantly elevated in the serum of HIV-positive individuals but did not adversely affect PPV23 response. Assessment of PPS-specific B cells revealed enhanced TACI and reduced BAFF-R expression compared to unselected B cells in HIV-positive and -negative individuals. Surface TACI was similar but soluble TACI was significantly lower in HIV-positive compared to HIV-negative individuals. Conclusion Current studies highlight a potential role for TACI in PPV23 response based on its enhanced expression on PPS-specific B cells. Although surface levels of TACI were similar, diminished soluble TACI (sTACI) in HIV-positive compared to HIV-negative individuals could potentially decrease BAFF responsiveness and Ig response. A better understanding of the role of TNF receptors could contribute to the design of improved pneumococcal vaccines. Trial Registration ClinicalTrials.gov NCT02515240
Collapse
Affiliation(s)
- Anita S Iyer
- Department of Medicine, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States of America
| | - Noor M Khaskhely
- Department of Medicine, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States of America
| | - David J Leggat
- Department of Medicine, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States of America
| | - Jennifer A Ohtola
- Department of Medicine, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States of America
| | - Jessica L Saul-McBeth
- Department of Medicine, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States of America
| | - Sadik A Khuder
- Department of Public Health, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States of America
| | - M A Julie Westerink
- Department of Medicine; Department of Infectious Diseases and Department of Microbiology and Immunology, 135 Rutledge Avenue, Charleston, South Carolina 29425, United States of America
| |
Collapse
|
81
|
Hohlfeld R, Dornmair K, Meinl E, Wekerle H. The search for the target antigens of multiple sclerosis, part 2: CD8+ T cells, B cells, and antibodies in the focus of reverse-translational research. Lancet Neurol 2015; 15:317-31. [PMID: 26724102 DOI: 10.1016/s1474-4422(15)00313-0] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 10/13/2015] [Accepted: 10/22/2015] [Indexed: 01/16/2023]
Abstract
Interest in CD8+ T cells and B cells was initially inspired by observations in multiple sclerosis rather than in animal models: CD8+ T cells predominate in multiple sclerosis lesions, oligoclonal immunoglobulin bands in CSF have long been recognised as diagnostic and prognostic markers, and anti-B-cell therapies showed considerable efficacy in multiple sclerosis. Taking a reverse-translational approach, findings from human T-cell receptor (TCR) and B-cell receptor (BCR) repertoire studies provided strong evidence for antigen-driven clonal expansion in the brain and CSF. New methods allow the reconstruction of human TCRs and antibodies from tissue-infiltrating immune cells, which can be used for the unbiased screening of antigen libraries. Myelin oligodendrocyte glycoprotein (MOG) has received renewed attention as an antibody target in childhood multiple sclerosis and in a small subgroup of adult patients with multiple sclerosis. Furthermore, there is growing evidence that a separate condition in adults exists, tentatively called MOG-antibody-associated encephalomyelitis, which has clinical features that overlap with neuromyelitis optica spectrum disorder and multiple sclerosis. Although CD8+ T cells and B cells are thought to have a pathogenic role in some subgroups of patients, their target antigens have yet to be identified.
Collapse
Affiliation(s)
- Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Campus Martinsried-Grosshadern, Ludwig-Maximilians University, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Campus Martinsried-Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Campus Martinsried-Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Hartmut Wekerle
- HERTIE Senior Professor Group Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany
| |
Collapse
|
82
|
Farjam M, Zhang GX, Ciric B, Rostami A. Emerging immunopharmacological targets in multiple sclerosis. J Neurol Sci 2015; 358:22-30. [PMID: 26440421 DOI: 10.1016/j.jns.2015.09.346] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 10/23/2022]
Abstract
Inflammatory demyelination of the central nervous system (CNS) is the hallmark of multiple sclerosis (MS), a chronic debilitating disease that affects more than 2.5 million individuals worldwide. It has been widely accepted, although not proven, that the major pathogenic mechanism of MS involves myelin-reactive T cell activation in the periphery and migration into the CNS, which subsequently triggers an inflammatory cascade that leads to demyelination and axonal damage. Virtually all MS medications now in use target the immune system and prevent tissue damage by modulating neuroinflammatory processes. Although current therapies such as commonly prescribed disease-modifying medications decrease the relapse rate in relapsing-remitting MS (RRMS), the prevention of long-term accumulation of deficits remains a challenge. Medications used for progressive forms of MS also have limited efficacy. The need for therapies that are effective against disease progression continues to drive the search for novel pharmacological targets. In recent years, due to a better understanding of MS immunopathogenesis, new approaches have been introduced that more specifically target autoreactive immune cells and their products, thus increasing specificity and efficacy, while reducing potential side effects such as global immunosuppression. In this review we describe several immunopharmacological targets that are currently being explored for MS therapy.
Collapse
Affiliation(s)
- Mojtaba Farjam
- Non-communicable Diseases Research Center, Department of Medical Pharmacology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
83
|
Goobie GC, Bernatsky S, Ramsey-Goldman R, Clarke AE. Malignancies in systemic lupus erythematosus: a 2015 update. Curr Opin Rheumatol 2015; 27:454-60. [PMID: 26125105 PMCID: PMC4562287 DOI: 10.1097/bor.0000000000000202] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Patients with systemic lupus erythematosus (SLE) have altered incidences of certain malignancies as compared with the general population. This review summarizes the recent literature on risk of malignancy in SLE and proposed mechanisms for these altered susceptibilities. RECENT FINDINGS Recent studies have confirmed previous data showing an increased risk of non-Hodgkin's lymphoma, lung, liver, vulvar/vaginal, and thyroid malignancies, whereas demonstrating a decreased risk of breast and prostate cancer. Lymphomagenesis in SLE has been linked to increased activity of multiple inflammatory cytokines as well as possible viral diseases. The decreased rates of hormone-sensitive cancers, such as breast and prostate, are speculated to be related to the presence of lupus autoantibodies and downregulation of certain proteins in SLE. This knowledge has been utilized to investigate new therapeutic modalities for these malignancies. SUMMARY Recent data confirm previously reported altered malignancy rates in SLE. Most striking in recent years are publications further elucidating mechanisms underlying cancer development in SLE, and subsequent investigations of potential therapeutics modulating these pathways.
Collapse
Affiliation(s)
- Gillian C Goobie
- Department of Medicine, Cumming School of Medicine, Health Sciences Centre, Foothills Campus, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N-4N1. . Phone: 403-710-9242
| | - Sasha Bernatsky
- Departments of Medicine and of Epidemiology and Biostatistics, McGill University, Montreal, Quebec, Canada
| | - Rosalind Ramsey-Goldman
- Department of Medicine, Division of Rheumatology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Ann E. Clarke
- Department of Medicine, Division of Rheumatology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
84
|
Abstract
While over the past decades T cells have been considered key players in the pathogenesis of multiple sclerosis (MS), it has only recently become evident that B cells have a major contributing role. Our understanding of the role of B cells has evolved substantially following the clinical success of B cell-targeting therapies and increasing experimental evidence for significant B cell involvement. Rather than mere antibody-producing cells, it is becoming clear that they are team players with the capacity to prime and regulate T cells, and function both as pro- and anti-inflammatory mediators. However, despite tremendous efforts, the target antigen(s) of B cells in MS have yet to be identified. The first part of this review summarizes the clinical evidence and results from animal studies pointing to the relevance of B cells in the pathogenesis of MS. The second part gives an overview of the currently known potential autoantigen targets. The third part recapitulates and critically appraises the currently available B cell-directed therapies.
Collapse
|
85
|
γ-Secretase directly sheds the survival receptor BCMA from plasma cells. Nat Commun 2015; 6:7333. [PMID: 26065893 PMCID: PMC4490565 DOI: 10.1038/ncomms8333] [Citation(s) in RCA: 256] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/28/2015] [Indexed: 12/26/2022] Open
Abstract
Survival of plasma cells is regulated by B-cell maturation antigen (BCMA), a membrane-bound receptor activated by its agonist ligands BAFF and APRIL. Here we report that γ-secretase directly cleaves BCMA, without prior truncation by another protease. This direct shedding is facilitated by the short length of BCMA's extracellular domain. In vitro, γ-secretase reduces BCMA-mediated NF-κB activation. In addition, γ-secretase releases soluble BCMA (sBCMA) that acts as a decoy neutralizing APRIL. In vivo, inhibition of γ-secretase enhances BCMA surface expression in plasma cells and increases their number in the bone marrow. Furthermore, in multiple sclerosis, sBCMA levels in spinal fluid are elevated and associated with intracerebral IgG production; in systemic lupus erythematosus, sBCMA levels in serum are elevated and correlate with disease activity. Together, shedding of BCMA by γ-secretase controls plasma cells in the bone marrow and yields a potential biomarker for B-cell involvement in human autoimmune diseases. B-cell maturation antigen (BCMA) regulates the survival of B cells and is essential for the maintenance of long-lived plasma cells. Here, the authors show that γ-secretase directly sheds BCMA from the cell surface and therefore regulates the number of plasma cells.
Collapse
|
86
|
Naradikian MS, Perate AR, Cancro MP. BAFF receptors and ligands create independent homeostatic niches for B cell subsets. Curr Opin Immunol 2015; 34:126-9. [PMID: 25836418 DOI: 10.1016/j.coi.2015.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 03/09/2015] [Accepted: 03/10/2015] [Indexed: 12/26/2022]
Abstract
The BAFF family of receptors and ligands controls B cell homeostasis and selection. Recent studies reveal distinct sources and roles for systemic versus locally produced BAFF. Moreover, the notion that differential BAFF receptor expression patterns establish independent homeostatic and selective niches has been strengthened. Finally, unique roles for BAFF family members in the regulation of antigen experienced and innate B cell subsets have been revealed. Herein, we overview current knowledge in these areas, emphasizing recent findings that inform these ideas.
Collapse
Affiliation(s)
- Martin S Naradikian
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.
| | - Alison R Perate
- Department of Anesthesiology and Critical Care, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Michael P Cancro
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
87
|
Effect of TACI signaling on humoral immunity and autoimmune diseases. J Immunol Res 2015; 2015:247426. [PMID: 25866827 PMCID: PMC4381970 DOI: 10.1155/2015/247426] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/04/2015] [Indexed: 02/02/2023] Open
Abstract
Transmembrane activator and calcium-modulating cyclophilin ligand interactor (TACI) is one of the receptors of B cell activating factor of the tumor necrosis factor family (BAFF) and a proliferation-inducing ligand (APRIL). TACI is a regulator in the immune responses. TACI inhibits B cell expansion and promotes the differentiation and survival of plasma cells. The mechanisms underlying these effects probably involve changed expressions of some crucial molecules, such as B lymphocyte induced maturation protein-1 (Blimp-1) and inducible T-cell costimulator ligand (ICOSL) in B cells and/or plasma cells. However, abnormal TACI signaling may relate to autoimmune disorders. Common variable immune deficiency (CVID) patients with heterozygous mutations in TACI alleles increase susceptibility to autoimmune diseases. Taci−/− mice and BAFF transgenic mice both develop signs of human SLE. These findings that indicate inappropriate levels of TACI signaling may disrupt immune system balance, thereby promoting the development of autoimmune diseases. In this review, we summarize the basic characteristics of the TACI ligands BAFF and APRIL, and detail the research findings on the role of TACI in humoral immunity. We also discuss the possible mechanisms underlying the susceptibility of CVID patients with TACI mutations to autoimmune diseases and the role of TACI in the pathogenesis of SLE.
Collapse
|