1
|
Li L, Li D, Jin J, Xu F, He N, Ren Y, Wang X, Tian L, Chen B, Li X, Chen Z, Zhang L, Qiao L, Wang L, Wang J. FOSL1-mediated LINC01566 negatively regulates CD4 + T-cell activation in myasthenia gravis. J Neuroinflammation 2024; 21:197. [PMID: 39113081 PMCID: PMC11308467 DOI: 10.1186/s12974-024-03194-5] [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: 04/18/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Myasthenia gravis (MG) is an autoimmune disease characterized by pathogenic antibodies that target structures of the neuromuscular junction. The evidence suggests that the regulation of long noncoding RNAs (lncRNAs) that is mediated by transcription factors (TFs) plays a key role in the pathophysiology of MG. Nevertheless, the detailed molecular mechanisms of lncRNAs in MG remain largely undetermined. METHODS Using microarray analysis, we analyzed the lncRNA levels in MG. By bioinformatics analysis, LINC01566 was found to potentially play an important role in MG. First, qRT‒PCR was performed to verify the LINC1566 expressions in MG patients. Then, fluorescence in situ hybridization was conducted to determine the localization of LINC01566 in CD4 + T cells. Finally, the impact of LINC01566 knockdown or overexpression on CD4 + T-cell function was also analyzed using flow cytometry and CCK-8 assay. A dual-luciferase reporter assay was used to validate the binding of the TF FOSL1 to the LINC01566 promoter. RESULTS Based on the lncRNA microarray and differential expression analyses, we identified 563 differentially expressed (DE) lncRNAs, 450 DE mRNAs and 19 DE TFs in MG. We then constructed a lncRNA-TF-mRNA network. Through network analysis, we found that LINC01566 may play a crucial role in MG by regulating T-cell-related pathways. Further experiments indicated that LINC01566 is expressed at low levels in MG patients. Functionally, LINC01566 is primarily distributed in the nucleus and can facilitate CD4 + T-cell apoptosis and inhibit cell proliferation. Mechanistically, we hypothesized that LINC01566 may negatively regulate the expressions of DUSP3, CCR2, FADD, SIRPB1, LGALS3 and SIRPB1, which are involved in the T-cell activation pathway, to further influence the cellular proliferation and apoptosis in MG. Moreover, we found that the effect of LINC01566 on CD4 + T cells in MG was mediated by the TF FOSL1, and in vitro experiments indicated that FOSL1 can bind to the promoter region of LINC01566. CONCLUSIONS In summary, our research revealed the protective roles of LINC01566 in clinical samples and cellular experiments, illustrating the potential roles and mechanism by which FOSL1/LINC01566 negatively regulates CD4 + T-cell activation in MG.
Collapse
Affiliation(s)
- Lifang Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Danyang Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Jingnan Jin
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Fanfan Xu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Ni He
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Yingjie Ren
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Xiaokun Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Liting Tian
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Biying Chen
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Xiaoju Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Zihong Chen
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Lanxin Zhang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Lukuan Qiao
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China.
| | - Jianjian Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
2
|
Watson SA, Javanmardi Y, Zanieri L, Shahreza S, Ragazzini R, Bonfanti P, Moeendarbary E. Integrated role of human thymic stromal cells in hematopoietic stem cell extravasation. Bioeng Transl Med 2023; 8:e10454. [PMID: 36925684 PMCID: PMC10013751 DOI: 10.1002/btm2.10454] [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: 06/10/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
The human thymus is the site of T-cell maturation and induction of central tolerance. Hematopoietic stem cell (HSC)-derived progenitors are recruited to the thymus from the fetal liver during early prenatal development and from bone marrow at later stages and postnatal life. The mechanism by which HSCs are recruited to the thymus is poorly understood in humans, though mouse models have indicated the critical role of thymic stromal cells (TSC). Here, we developed a 3D microfluidic assay based on human cells to model HSC extravasation across the endothelium into the extracellular matrix. We found that the presence of human TSC consisting of cultured thymic epithelial cells (TEC) and interstitial cells (TIC) increases the HSC extravasation rates by 3-fold. Strikingly, incorporating TEC or TIC alone is insufficient to perturb HSC extravasation rates. Furthermore, we identified complex gene expressions from interactions between endothelial cells, TEC and TIC modulates the HSCs extravasation. Our results suggest that comprehensive signaling from the complex thymic microenvironment is crucial for thymus seeding and that our system will allow manipulation of these signals with the potential to increase thymocyte migration in a therapeutic setting.
Collapse
Affiliation(s)
- Sara A. Watson
- Department of Mechanical EngineeringUCLLondonUK
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
| | | | - Luca Zanieri
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | | | - Roberta Ragazzini
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | - Paola Bonfanti
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | | |
Collapse
|
3
|
Donor natural killer cells trigger production of β-2-microglobulin to enhance post-bone marrow transplant immunity. Blood 2022; 140:2323-2334. [PMID: 35984965 DOI: 10.1182/blood.2021015297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 07/14/2022] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic transplantation is a powerful treatment for hematologic malignancies. Posttransplant immune incompetence exposes patients to disease relapse and infections. We previously demonstrated that donor alloreactive natural killer (NK) cells ablate recipient hematopoietic targets, including leukemia. Here, in murine models, we show that infusion of donor alloreactive NK cells triggers recipient dendritic cells (DCs) to synthesize β-2-microglobulin (B2M) that elicits the release of c-KIT ligand and interleukin-7 that greatly accelerate posttransplant immune reconstitution. An identical chain of events was reproduced by infusing supernatants of alloreactive NK/DC cocultures. Similarly, human alloreactive NK cells triggered human DCs to synthesize B2M that induced interleukin-7 production by thymic epithelial cells and thereby supported thymocyte cellularity in vitro. Chromatography fractionation of murine and human alloreactive NK/DC coculture supernatants identified a protein with molecular weight and isoelectric point of B2M, and mass spectrometry identified amino acid sequences specific of B2M. Anti-B2M antibody depletion of NK/DC coculture supernatants abrogated their immune-rebuilding effect. B2M knock-out mice were unable to undergo accelerated immune reconstitution, but infusion of (wild-type) NK/DC coculture supernatants restored their ability to undergo accelerated immune reconstitution. Similarly, silencing the B2M gene in human DCs, before coculture with alloreactive NK cells, prevented the increase in thymocyte cellularity in vitro. Finally, human recombinant B2M increased thymocyte cellularity in a thymic epithelial cells/thymocyte culture system. Our studies uncover a novel therapeutic principle for treating posttransplant immune incompetence and suggest that, upon its translation to the clinic, patients may benefit from adoptive transfer of large numbers of cytokine-activated, ex vivo-expanded donor alloreactive NK cells.
Collapse
|
4
|
Ma J, Gong Q, Pan X, Guo P, He L, You Y. Depletion of Fractalkine ameliorates renal injury and Treg cell apoptosis via the p38MAPK pathway in lupus-prone mice. Exp Cell Res 2021; 405:112704. [PMID: 34126056 DOI: 10.1016/j.yexcr.2021.112704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 11/20/2022]
Abstract
Fractalkine (FKN) is a chemokine with several roles, including chemotaxis; adhesion; and immune damage, which also participates in cell inflammation and apoptosis and responds to the pathogenesis of autoimmune diseases. Given the involvement of regulatory T cells (Treg) cells in autoimmune diseases, this study investigated the regulatory mechanism of FKN in renal injury and Treg apoptosis via the p38 mitogen-activated protein kinase (p38MAPK) signaling pathway in lupus-prone mice. Lupus was induced in BALB/c female mice by injection of pristane, followed by isolation of CD4+CD25+ Treg cells from the spleen of lupus model mice. To deplete FKN, mice received injection of an anti-FKN antibody, and Treg cells were transfected with FKN small-interfering RNA. Lupus mice and Treg cells were treated with the p38MAPK inhibitor SB203580 and activator U-46619, respectively, and urine protein and serum urea nitrogen, creatinine, and autoantibodies were measured and renal histopathological changes analyzed. We determined levels of FKN, phosphorylated p38 (p-p38), and forkhead box P3 (FOXP3) in renal tissue and Treg cells, and analyzed apoptosis rates and levels of key apoptotic factors in Treg cells. The renal FKN and p-p38 levels increased, whereas renal FOXP3 level decreased in lupus-prone mice. Treatment with the anti-FKN antibody and the p38MAPK inhibitor ameliorated proteinuria and renal function, significantly reducing serum autoantibody, renal FKN, and p-p38 levels while increasing renal FOXP3 level in lupus-prone mice. Moreover, FKN knockdown and administration of the p38MAPK inhibitor reduced apoptosis and levels of pro-apoptotic factors, increased levels of anti-apoptotic factors, and suppressed activation of p38MAPK signaling in Treg cells derived from lupus model mice. Furthermore, treatment with the p38MAPK activator U-46619 had the opposite effect on these cells. These data indicated that depletion of FKN ameliorated renal injury and Treg cell apoptosis via inhibition of p38MAPK signaling in lupus nephritis, suggesting that targeting FKN represents a potential therapeutic strategy for treating Lupus nephritis.
Collapse
Affiliation(s)
- Jingxue Ma
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Qiming Gong
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Xiuhong Pan
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Pengwei Guo
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Linlin He
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Yanwu You
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
5
|
Selective involution of thymic medulla by cyclosporine A with a decrease of mature thymic epithelia, XCR1 + dendritic cells, and epithelium-free areas containing Foxp3 + thymic regulatory T cells. Histochem Cell Biol 2021; 156:133-146. [PMID: 33993340 PMCID: PMC8397693 DOI: 10.1007/s00418-021-01993-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2021] [Indexed: 11/25/2022]
Abstract
Immunosuppressive drugs such as cyclosporine A (CSA) can disrupt thymic structure and functions, ultimately inducing syngeneic/autologous graft-versus-host disease together with involuted medullas. To elucidate the effects of CSA on the thymus more precisely, we analyzed the effects of CSA on the thymus and T cell system using rats. In addition to confirming the phenomena already reported, we newly found that the proportion of recent thymic emigrants also greatly decreased, suggesting impaired supply. Immunohistologically, the medullary thymic epithelial cells (mTECs) presented with a relative decrease in the subset with a competent phenotype and downregulation of class II major histocompatibility complex molecules. In control rats, thymic dendritic cells (DCs) comprised two subsets, XCR1+SIRP1α−CD4− and XCR1−SIRP1α+CD4+. The former had a tendency to selectively localize in the previously-reported epithelium-containing areas of the rat medullas, and the number was significantly reduced by CSA treatment. The epithelium-free areas, another unique domains in the rat medullas, contained significantly more Foxp3+ thymic Tregs. With CSA treatment, the epithelium-free areas presented strong involution, and the number and distribution of Tregs in the medulla were greatly reduced. These results suggest that CSA inhibits the production of single-positive thymocytes, including Tregs, and disturbs the microenvironment of the thymic medulla, with a decrease of the competent mTECs and disorganization of epithelium-free areas and DC subsets, leading to a generation of autoreactive T cells with selective medullary involution.
Collapse
|
6
|
Truffault F, Nazzal D, Verdier J, Gradolatto A, Fadel E, Roussin R, Eymard B, Le Panse R, Berrih-Aknin S. Comparative Analysis of Thymic and Blood Treg in Myasthenia Gravis: Thymic Epithelial Cells Contribute to Thymic Immunoregulatory Defects. Front Immunol 2020; 11:782. [PMID: 32435245 PMCID: PMC7218102 DOI: 10.3389/fimmu.2020.00782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/06/2020] [Indexed: 11/13/2022] Open
Abstract
The thymus is involved in autoimmune Myasthenia gravis (MG) associated with anti-acetylcholine (AChR) antibodies. In MG, thymic regulatory T cells (Treg) are not efficiently suppressive, and conventional T cells (Tconv) are resistant to suppression. To better understand the specific role of the thymus in MG, we compared the phenotype and function of peripheral and thymic Treg and Tconv from controls and MG patients. Suppression assays with thymic or peripheral CD4 + T cells showed that the functional impairment in MG was more pronounced in the thymus than in the periphery. Phenotypic analysis of Treg showed a significant reduction of resting and effector Treg in the thymus but not in the periphery of MG patients. CD31, a marker lost with excessive immunoreactivity, was significantly reduced in thymic but not blood resting Treg. These results suggest that an altered thymic environment may explain Treg differences between MG patients and controls. Since thymic epithelial cells (TECs) play a major role in the generation of Treg, we co-cultured healthy thymic CD4 + T cells with control or MG TECs and tested their suppressive function. Co-culture with MG TECs consistently hampers regulatory activity, as compared with control TECs, suggesting that MG TECs contribute to the immune regulation defects of MG CD4 + T cells. MG TECs produced significantly higher thymic stromal lymphopoietin (TSLP) than control TECs, and a neutralizing anti-TSLP antibody partially restored the suppressive capacity of Treg derived from co-cultures with MG TECs, suggesting that TSLP contributed to the defect of thymic Treg in MG patients. Finally, a co-culture of MG CD4 + T cells with control TECs restored numbers and function of MG Treg, demonstrating that a favorable environment could correct the immune regulation defects of T cells in MG. Altogether, our data suggest that the severe defect of thymic Treg is at least partially due to MG TECs that overproduce TSLP. The Treg defects could be corrected by replacing dysfunctional TECs by healthy TECs. These findings highlight the role of the tissue environment on the immune regulation.
Collapse
Affiliation(s)
- Frédérique Truffault
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Dani Nazzal
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Julien Verdier
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Angeline Gradolatto
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Elie Fadel
- Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | | | - Bruno Eymard
- AP-HP, Referral Center for Neuromuscular Disorders, Pitié-Salpêtrière Hospital, Institute of Myology, Paris, France
| | - Rozen Le Panse
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| |
Collapse
|
7
|
Villegas JA, Bayer AC, Ider K, Bismuth J, Truffault F, Roussin R, Santelmo N, Le Panse R, Berrih-Aknin S, Dragin N. Il-23/Th17 cell pathway: A promising target to alleviate thymic inflammation maintenance in myasthenia gravis. J Autoimmun 2019; 98:59-73. [PMID: 30578016 DOI: 10.1016/j.jaut.2018.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/27/2022]
Abstract
IL-23/Th17 pathway has been identified to sustain inflammatory condition in several autoimmune diseases and therefore being targeted in various therapeutic and effective approaches. Patients affected with autoimmune myasthenia gravis exhibit a disease effector tissue, the thymus, that harbors ectopic germinal centers that sustain production of auto-antibodies, targeting proteins located in the neuromuscular junction, cause of the organ-specific chronic autoimmune disease. The present study aims to investigate the IL-23/Th17 cell pathway in the thymic inflammatory and pathogenic events. We found that thymuses of MG patients displayed overexpression of Interleukin-17, signature cytokine of activated Th17 cells. This activation was sustained by a higher secretion of Interleukin-23 by TEC, in addition to the increased expression of cytokines involved in Th17 cell development. The overexpression of Interleukin-23 was due to a dysregulation of interferon type I pathway. Besides, Interleukin-17 secreted, and Th17 cells were localized around thymic ectopic germinal centers. These cells expressed podoplanin, a protein involved in B-cell maturation and antibody secretion. Finally, production of Interleukin-23 was also promoted by Interleukin-17 secreted itself by Th17 cells, highlighting a chronic loop of inflammation sustained by thymic cell interaction. Activation of the IL-23/Th17 pathway in the thymus of autoimmune myasthenia gravis patients creates an unstoppable loop of inflammation that may participate in ectopic germinal center maintenance. To alleviate the physio-pathological events in myasthenia gravis patients, this pathway may be considered as a new therapeutic target.
Collapse
Affiliation(s)
- José A Villegas
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Alexandra C Bayer
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Katia Ider
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Jacky Bismuth
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Frédérique Truffault
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | | | | | - Rozen Le Panse
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France.
| | - Sonia Berrih-Aknin
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Nadine Dragin
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Inovarion, Paris, France.
| |
Collapse
|
8
|
Cron MA, Maillard S, Villegas J, Truffault F, Sudres M, Dragin N, Berrih-Aknin S, Le Panse R. Thymus involvement in early-onset myasthenia gravis. Ann N Y Acad Sci 2017; 1412:137-145. [DOI: 10.1111/nyas.13519] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/05/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Mélanie A. Cron
- INSERM U974; Paris France
- UPMC Sorbonne Universités; Paris France
- AIM; Institut de myologie; Paris France
| | - Solène Maillard
- INSERM U974; Paris France
- UPMC Sorbonne Universités; Paris France
- AIM; Institut de myologie; Paris France
| | - José Villegas
- INSERM U974; Paris France
- UPMC Sorbonne Universités; Paris France
- AIM; Institut de myologie; Paris France
| | - Frédérique Truffault
- INSERM U974; Paris France
- UPMC Sorbonne Universités; Paris France
- AIM; Institut de myologie; Paris France
| | - Muriel Sudres
- INSERM U974; Paris France
- UPMC Sorbonne Universités; Paris France
- AIM; Institut de myologie; Paris France
| | - Nadine Dragin
- INSERM U974; Paris France
- UPMC Sorbonne Universités; Paris France
- AIM; Institut de myologie; Paris France
| | - Sonia Berrih-Aknin
- INSERM U974; Paris France
- UPMC Sorbonne Universités; Paris France
- AIM; Institut de myologie; Paris France
| | - Rozen Le Panse
- INSERM U974; Paris France
- UPMC Sorbonne Universités; Paris France
- AIM; Institut de myologie; Paris France
| |
Collapse
|
9
|
Response to: Comment on "Detecting Key Genes Regulated by miRNAs in Dysfunctional Crosstalk Pathway of Myasthenia Gravis". BIOMED RESEARCH INTERNATIONAL 2017; 2017:5359434. [PMID: 28884124 PMCID: PMC5573091 DOI: 10.1155/2017/5359434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/20/2017] [Indexed: 11/17/2022]
|
10
|
Losada-Barragán M, Umaña-Pérez A, Cuervo-Escobar S, Berbert LR, Porrozzi R, Morgado FN, Mendes-da-Cruz DA, Savino W, Sánchez-Gómez M, Cuervo P. Protein malnutrition promotes dysregulation of molecules involved in T cell migration in the thymus of mice infected with Leishmania infantum. Sci Rep 2017; 7:45991. [PMID: 28397794 PMCID: PMC5387407 DOI: 10.1038/srep45991] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/07/2017] [Indexed: 12/18/2022] Open
Abstract
Protein malnutrition, the most deleterious cause of malnutrition in developing countries, has been considered a primary risk factor for the development of clinical visceral leishmaniasis (VL). Protein malnutrition and infection with Leishmania infantum leads to lymphoid tissue disorganization, including changes in cellularity and lymphocyte subpopulations in the thymus and spleen. Here we report that protein malnutrition modifies thymic chemotactic factors by diminishing the CCL5, CXCL12, IGF1, CXCL9 and CXCL10 protein levels in infected animals. Nevertheless, T cells preserve their migratory capability, as they were able to migrate ex vivo in response to chemotactic stimuli, indicating that malnutrition may compromise the thymic microenvironment and alter in vivo thymocyte migration. Decrease in chemotactic factors protein levels was accompanied by an early increase in the parasite load of the spleen. These results suggest that the precondition of malnutrition is affecting the cell-mediated immune response to L. infantum by altering T cell migration and interfering with the capacity of protein-deprived animals to control parasite spreading and proliferation. Our data provide evidence for a disturbance of T lymphocyte migration involving both central and peripheral T-cells, which likely contribute to the pathophysiology of VL that occurs in malnourished individuals.
Collapse
Affiliation(s)
- Monica Losada-Barragán
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Adriana Umaña-Pérez
- Departamento de Química, Universidad Nacional de Colombia, Sede Bogotá, Facultad de Ciencias, Grupo de Investigación en Hormonas, Bogotá, Colombia
| | - Sergio Cuervo-Escobar
- Departamento de Química, Universidad Nacional de Colombia, Sede Bogotá, Facultad de Ciencias, Grupo de Investigación en Hormonas, Bogotá, Colombia
| | - Luiz Ricardo Berbert
- Laboratório de Pesquisas sobre o Timo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Renato Porrozzi
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Fernanda N Morgado
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | | | - Wilson Savino
- Laboratório de Pesquisas sobre o Timo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Myriam Sánchez-Gómez
- Departamento de Química, Universidad Nacional de Colombia, Sede Bogotá, Facultad de Ciencias, Grupo de Investigación en Hormonas, Bogotá, Colombia.
| | - Patricia Cuervo
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil.
| |
Collapse
|
11
|
Pathological Findings in Myasthenia Gravis Patients with Thymic Hyperplasia and Thymoma. Pathol Oncol Res 2017; 24:67-74. [DOI: 10.1007/s12253-017-0213-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 02/16/2017] [Indexed: 12/28/2022]
|
12
|
Truffault F, de Montpreville V, Eymard B, Sharshar T, Le Panse R, Berrih-Aknin S. Thymic Germinal Centers and Corticosteroids in Myasthenia Gravis: an Immunopathological Study in 1035 Cases and a Critical Review. Clin Rev Allergy Immunol 2017; 52:108-124. [PMID: 27273086 DOI: 10.1007/s12016-016-8558-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The most common form of Myasthenia gravis (MG) is due to anti-acetylcholine receptor (AChR) antibodies and is frequently associated with thymic pathology. In this review, we discuss the immunopathological characteristics and molecular mechanisms of thymic follicular hyperplasia, the effects of corticosteroids on this thymic pathology, and the role of thymic epithelial cells (TEC), a key player in the inflammatory thymic mechanisms. This review is based not only on the literature data but also on thymic transcriptome results and analyses of pathological and immunological correlations in a vast cohort of 1035 MG patients without thymoma. We show that among patients presenting a thymic hyperplasia with germinal centers (GC), 80 % are females, indicating that thymic follicular hyperplasia is mainly a disease of women. The presence of anti-AChR antibodies is correlated with the degree of follicular hyperplasia, suggesting that the thymus is a source of anti-AChR antibodies. The degree of hyperplasia is not dependent upon the time from the onset, implying that either the antigen is chronically expressed and/or that the mechanisms of the resolution of the GC are not efficiently controlled. Glucocorticoids, a conventional therapy in MG, induce a significant reduction in the GC number, together with changes in the expression of chemokines and angiogenesis. These changes are likely related to the acetylation molecular process, overrepresented in corticosteroid-treated patients, and essential for gene regulation. Altogether, based on the pathological and molecular thymic abnormalities found in MG patients, this review provides some explanations for the benefit of thymectomy in early-onset MG patients.
Collapse
Affiliation(s)
- Frédérique Truffault
- INSERM U974, Paris, France.,CNRS FRE3617, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,AIM, Institut de myologie, Paris, France
| | | | - Bruno Eymard
- Department of Neuromuscular Disorders, CHU Salpêtrière, Paris, France
| | - Tarek Sharshar
- General Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Raymond Poincaré Hospital, University of Versailles Saint-Quentin en Yvelines, 92380, Garches, France
| | - Rozen Le Panse
- INSERM U974, Paris, France.,CNRS FRE3617, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,AIM, Institut de myologie, Paris, France
| | - Sonia Berrih-Aknin
- INSERM U974, Paris, France. .,CNRS FRE3617, Paris, France. .,Sorbonne Universités, UPMC Univ Paris 06, Paris, France. .,AIM, Institut de myologie, Paris, France. .,UMRS 974 UPMC, INSERM, FRE 3617 CNRS, AIM, Center of Research in Myology, 105 Boulevard de l'Hôpital, Paris, 75013, France.
| |
Collapse
|
13
|
Feng H, Pyykkö I, Zou J. Involvement of Ubiquitin-Editing Protein A20 in Modulating Inflammation in Rat Cochlea Associated with Silver Nanoparticle-Induced CD68 Upregulation and TLR4 Activation. NANOSCALE RESEARCH LETTERS 2016; 11:240. [PMID: 27142878 PMCID: PMC4854861 DOI: 10.1186/s11671-016-1430-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/13/2016] [Indexed: 06/05/2023]
Abstract
Silver nanoparticles (AgNPs) were shown to temporarily impair the biological barriers in the skin of the external ear canal, mucosa of the middle ear, and inner ear, causing partially reversible hearing loss after delivery into the middle ear. The current study aimed to elucidate the molecular mechanism, emphasizing the TLR signaling pathways in association with the potential recruitment of macrophages in the cochlea and the modulation of inflammation by ubiquitin-editing protein A20. Molecules potentially involved in these signaling pathways were thoroughly analysed using immunohistochemistry in the rat cochlea exposed to AgNPs at various concentrations through intratympanic injection. The results showed that 0.4 % AgNPs but not 0.02 % AgNPs upregulated the expressions of CD68, TLR4, MCP1, A20, and RNF11 in the strial basal cells, spiral ligament fibrocytes, and non-sensory supporting cells of Corti's organ. 0.4 % AgNPs had no effect on CD44, TLR2, MCP2, Rac1, myosin light chain, VCAM1, Erk1/2, JNK, p38, IL-1β, TNF-α, TNFR1, TNFR2, IL-10, or TGF-β. This study suggested that AgNPs might confer macrophage-like functions on the strial basal cells and spiral ligament fibrocytes and enhance the immune activities of non-sensory supporting cells of Corti's organ through the upregulation of CD68, which might be involved in TLR4 activation. A20 and RNF11 played roles in maintaining cochlear homeostasis via negative regulation of the expressions of inflammatory cytokines.
Collapse
Affiliation(s)
- Hao Feng
- Hearing and Balance Research Unit, Field of Oto-laryngology, School of Medicine, University of Tampere, Medisiinarinkatu 3, 33520, Tampere, Finland
| | - Ilmari Pyykkö
- Hearing and Balance Research Unit, Field of Oto-laryngology, School of Medicine, University of Tampere, Medisiinarinkatu 3, 33520, Tampere, Finland
| | - Jing Zou
- Hearing and Balance Research Unit, Field of Oto-laryngology, School of Medicine, University of Tampere, Medisiinarinkatu 3, 33520, Tampere, Finland.
- Department of Otolaryngology-Head and Neck Surgery, Center for Otolaryngology-Head and Neck Surgery of Chinese PLA, Changhai Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
14
|
Akahane M, Watanabe M, Inoue N, Miyahara Y, Arakawa Y, Inoue Y, Katsumata Y, Hidaka Y, Iwatani Y. Association of the polymorphisms of chemokine genes (IL8, RANTES, MIG, IP10, MCP1 and IL16) with the pathogenesis of autoimmune thyroid diseases. Autoimmunity 2016; 49:312-9. [DOI: 10.3109/08916934.2015.1134507] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Maiko Akahane
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan and
| | - Mikio Watanabe
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan and
| | - Naoya Inoue
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan and
| | - Yumi Miyahara
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan and
| | - Yuya Arakawa
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan and
| | - Yuka Inoue
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan and
| | - Yuka Katsumata
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan and
| | - Yoh Hidaka
- Laboratory for Clinical Investigation, Osaka University Hospital, Osaka, Japan
| | - Yoshinori Iwatani
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan and
| |
Collapse
|
15
|
Systems biology of myasthenia gravis, integration of aberrant lncRNA and mRNA expression changes. BMC Med Genomics 2015; 8:13. [PMID: 25889429 PMCID: PMC4380247 DOI: 10.1186/s12920-015-0087-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 02/26/2015] [Indexed: 12/20/2022] Open
Abstract
Background A novel class of transcripts, long non-coding RNAs (lncRNAs), has recently emerged as a key player in several biological processes, and important roles for these molecules have been reported in a number of complex human diseases, such as autoimmune diseases, neurological disorders, and various cancers. However, the aberrant lncRNAs implicated in myasthenia gravis (MG) remain unknown. The aim of the present study was to explore the abnormal expression of lncRNAs in peripheral blood mononuclear cells (PBMCs) and examine mRNA regulatory relationship networks among MG patients with or without thymoma. Methods Microarray assays were performed, and the outstanding differences between lncRNAs or mRNA expression were verified through RT-PCR. The lncRNAs functions were annotated for the target genes using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) biological pathway. The potential regulatory relationships between the lncRNAs and target genes were analyzed using the ‘cis’ and ‘trans’ model. Outstanding lncRNAs were organized to generate a TF-lncRNA-gene network using Cytoscape software. Results The lncRNA and mRNA expression profile analysis revealed subsets of differentially expressed genes in MG patients with or without thymoma. A total of 12 outstanding dysregulated expression lncRNAs, such as lncRNA oebiotech_11933, were verified through real-time PCR. Several GO terms including the cellular response to interferon-γ, platelet degranulation, chemokine receptor binding and cytokine interactions were very important in MG pathogenesis. The chromosome locations of some lncRNAs and associated co-expression genes were demonstrated using ‘cis’ analysis. The results of the ‘trans’ analysis revealed that some TFs (i.e., CTCF, TAF1and MYC) regulate lncRNA and gene expression. The outstanding lncRNAs in each group were implicated in the regulation of the TF-lncRNA-target gene network. Conclusion The results of the present study provide a perspective on lncRNA expression in MG. We identify a subset of aberrant lncRNAs and mRNAs as potential biomarkers for the diagnosis of MG. The GO and KEGG pathway analysis provides an annotation to determine the functions of these lncRNAs. The results of the ‘cis’ and ‘trans’ analyses provide information concerning the modular regulation of lncRNAs. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0087-z) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Detecting key genes regulated by miRNAs in dysfunctional crosstalk pathway of myasthenia gravis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:724715. [PMID: 25705681 PMCID: PMC4331476 DOI: 10.1155/2015/724715] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/10/2014] [Indexed: 02/07/2023]
Abstract
Myasthenia gravis (MG) is a neuromuscular autoimmune disorder resulting from autoantibodies attacking components of the neuromuscular junction. Recent studies have implicated the aberrant expression of microRNAs (miRNAs) in the pathogenesis of MG; however, the underlying mechanisms remain largely unknown. This study aimed to identify key genes regulated by miRNAs in MG. Six dysregulated pathways were identified through differentially expressed miRNAs and mRNAs in MG, and significant crosstalk was detected between five of these. Notably, crosstalk between the "synaptic long-term potentiation" pathway and four others was mediated by five genes involved in the MAPK signaling pathway. Furthermore, 14 key genes regulated by miRNAs were detected, of which six-MAPK1, RAF1, PGF, PDGFRA, EP300, and PPP1CC-mediated interactions between the dysregulated pathways. MAPK1 and RAF1 were responsible for most of this crosstalk (80%), likely reflecting their central roles in MG pathogenesis. In addition, most key genes were enriched in immune-related local areas that were strongly disordered in MG. These results provide new insight into the pathogenesis of MG and offer new potential targets for therapeutic intervention.
Collapse
|
17
|
Yang L, Wang J, Sun X, Cao Y, Ning S, Zhang H, Chen L, Li R, Tian Q, Wang L, Wang W, Li X. Identifying a polymorphic 'switch' that influences miRNAs' regulation of a myasthenia gravis risk pathway. PLoS One 2014; 9:e104827. [PMID: 25118158 PMCID: PMC4130595 DOI: 10.1371/journal.pone.0104827] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 07/18/2014] [Indexed: 12/03/2022] Open
Abstract
The significant roles of genetic variants in myasthenia gravis (MG) pathogenesis have been demonstrated in many studies, and recently it has been revealed that aberrant level/regulation of microRNAs (miRNAs) might contribute to the initiation and progression of MG. However, the dysfunction of miRNA associated with single nucleotide polymorphisms (miRSNPs) has not been well investigated in MG. In this study, we created a contemporary catalog of 89 MG risk genes via manual literature-mining. Based on this risk gene catalog, we obtained 18 MG risk pathways. Furthermore, we identified 93 miRNAs that target MG risk pathways and revealed the miRSNPs ‘switches’ in miRNA regulation in the MG risk pathways by integrating the database information of miRSNPs. We also constructed a miRNA-mediated SNP switching pathway network (MSSPN) to intuitively analyze miRNA regulation of MG risk pathways and the relationship of the polymorphism ‘switch’ with these changes in regulation. Moreover, we carried out in-depth dissection on the correlation between hsa05200 (pathway in cancer) and MG development, and elaborated the significance of 4 high-risk genes. By network analysis and literature mining, we proposed a potential mechanism of miRSNPs→gene→pathway effects on MG pathogenesis, especially for rs28457673 (miR-15/16/195/424/497 family)→IGF1R→hsa05200 (pathway in cancer). Therefore, our studies have revealed a functional role for genetic modulators in MG pathogenesis at a systemic level, which could be informative for further miRNA and miRSNPs studies in MG.
Collapse
Affiliation(s)
- Lili Yang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jianjian Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xuesong Sun
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yuze Cao
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Huixue Zhang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lixia Chen
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ronghong Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qinghua Tian
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
- * E-mail: (LW); (WW); (XL)
| | - Weizhi Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang Province, China
- * E-mail: (LW); (WW); (XL)
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, China
- * E-mail: (LW); (WW); (XL)
| |
Collapse
|
18
|
Ferreira C, Palmer D, Blake K, Garden OA, Dyson J. Reduced regulatory T cell diversity in NOD mice is linked to early events in the thymus. THE JOURNAL OF IMMUNOLOGY 2014; 192:4145-52. [PMID: 24663675 DOI: 10.4049/jimmunol.1301600] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The thymic natural regulatory T cell (Treg) compartment of NOD mice is unusual in having reduced TCR diversity despite normal cellularity. In this study, we show that this phenotype is attributable to perturbations in early and late stages of thymocyte development and is controlled, at least in part, by the NOD Idd9 region on chromosome 4. Progression from double negative 1 to double negative 2 stage thymocytes in NOD mice is inefficient; however, this defect is compensated by increased proliferation of natural Tregs (nTregs) within the single positive CD4 thymocyte compartment, accounting for recovery of cellularity accompanied by loss of TCR diversity. This region also underlies the known attenuation of ERK-MAPK signaling, which may preferentially disadvantage nTreg selection. Interestingly, the same genetic region also regulates the rate of thymic involution that is accelerated in NOD mice. These findings highlight further complexity in the control of nTreg repertoire diversity.
Collapse
Affiliation(s)
- Cristina Ferreira
- Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | | | | | | | | |
Collapse
|
19
|
Weiss JM, Cufi P, Le Panse R, Berrih-Aknin S. The thymus in autoimmune Myasthenia Gravis: Paradigm for a tertiary lymphoid organ. Rev Neurol (Paris) 2013; 169:640-9. [DOI: 10.1016/j.neurol.2013.02.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 01/10/2013] [Accepted: 02/04/2013] [Indexed: 01/02/2023]
|
20
|
Kiaii S, Kokhaei P, Mozaffari F, Rossmann E, Pak F, Moshfegh A, Palma M, Hansson L, Mashayekhi K, Hojjat-Farsangi M, Österborg A, Choudhury A, Mellstedt H. T cells from indolent CLL patients prevent apoptosis of leukemic B cells in vitro and have altered gene expression profile. Cancer Immunol Immunother 2013; 62:51-63. [PMID: 22736254 PMCID: PMC11029037 DOI: 10.1007/s00262-012-1300-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 05/30/2012] [Indexed: 10/28/2022]
Abstract
T cells may have a role in sustaining the leukemic clone in chronic lymphocytic leukemia (CLL). In this study, we have examined the ability of T cells from CLL patients to support the survival of the leukemic B cells in vitro. Additionally, we compared global gene expression of T cells from indolent CLL patients with healthy individuals and multiple myeloma (MM) patients. Apoptosis of purified leukemic B cells was inhibited in vitro when co-cultured with increasing numbers of autologous T cells (p < 0.01) but not autologous B and T cells of normal donors. The anti-apoptotic effect exceeded that of the anti-apoptotic cytokine IL-4 (p = 0.002) and was greater with CD8+ cells (p = 0.02) than with CD4+ cells (p = 0.05). The effect was depended mainly on cell-cell contact although a significant effect was also observed in transwell experiments (p = 0.05). About 356 genes involved in different cellular pathways were deregulated in T cells of CLL patients compared to healthy individuals and MM patients. The results of gene expression profiling were verified for 6 genes (CCL4, CCL5 (RANTES), XCL1, XCL2, KLF6, and TRAF1) using qRT-PCR and immunoblotting. Our results demonstrate that CLL-derived T cells can prevent apoptosis of leukemic B cells and have altered expression of genes that may facilitate the survival of the leukemic clone.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Apoptosis/immunology
- B-Lymphocytes/immunology
- Coculture Techniques
- Female
- Flow Cytometry
- Humans
- Immunoblotting
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Middle Aged
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes/immunology
- Transcriptome
Collapse
Affiliation(s)
- Shahryar Kiaii
- Institute of Cancer, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London, EC1M 6BQ UK
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Parviz Kokhaei
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Fariba Mozaffari
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Eva Rossmann
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Fatemeh Pak
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Ali Moshfegh
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Marzia Palma
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Lotta Hansson
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Kaveh Mashayekhi
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Mohammad Hojjat-Farsangi
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Anders Österborg
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Aniruddha Choudhury
- Center for Immune and Targeted Therapy Greenslopes Private Hospital, Newdegate Street, Brisbane, QLD 4072 Australia
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Håkan Mellstedt
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| |
Collapse
|
21
|
Zhi-Chun L, Qiao-Ling Z, Zhi-Qin L, Xiao-Zhao L, Xiao-xia Z, Rong T. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) mediates p38 mitogen-activated protein kinase activation and signal transduction in peripheral blood mononuclear cells from patients with lupus nephritis. Inflammation 2012; 35:935-43. [PMID: 22009442 DOI: 10.1007/s10753-011-9396-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Forty-two patients with systemic lupus erythematosus (SLE), including 26 patients with renal damage and 16 without, and 20 healthy controls were included in the study. The isolated peripheral blood mononuclear cells (PBMCs) were treated with a p38 inhibitor (SB203580) or anti-tumor necrosis factor-like weak inducer of apoptosis (TWEAK) mAb, with or without phytohemagglutinin/phorbol myristate acetate (PHA/PMA) stimulation. Western blot experiments were used to evaluate the protein expression of TWEAK and p38 MAPK in PBMCs .Next, the contents of interleukin-10 (IL-10) and monocyte chemoattractant protein-1 (MCP-1) in the supernatant were measured by ELISA. The results showed that expression of TWEAK protein in PBMCs from lupus nephritis patients was significantly higher than that from SLE patients without renal damage and healthy controls. PHA/PMA simulation could upregulate the productions of TWEAK and p-p38MAPK in PBMCs from patients with SLE. Anti-TWEAK mAb treatment downregulated both TWEAK and p-p38 MAPK expression in PBMCs, as well as IL-10 and MCP-1 in the supernatant; SB203580 had the same effect on cytokine production in PBMC, but had no effect on the expression of TWEAK. Our results suggested that TWEAK-p38 MAPK-IL-10, MCP-1 signaling pathway in PBMC played an important pathogenic role in lupus nephritis.
Collapse
Affiliation(s)
- Liu Zhi-Chun
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Suzhou University, Suzhou, China
| | | | | | | | | | | |
Collapse
|
22
|
Jain G, Cronauer MV, Schrader M, Möller P, Marienfeld RB. NF-κB signaling in prostate cancer: a promising therapeutic target? World J Urol 2011; 30:303-10. [PMID: 22085980 DOI: 10.1007/s00345-011-0792-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 10/19/2011] [Indexed: 01/31/2023] Open
Abstract
Prostate carcinoma (PCa) displays a wide variety of genetic alterations, versatile expression profiles as well as cell surface markers. Despite this heterogeneity, a common treatment for advanced PCa is androgen deprivation therapy (ADT). ADT targets the androgen receptor-a member of the nuclear receptor superfamily-which is required for development and function of the prostate and critical for PCa growth and survival. After an initial regression of the tumor during ADT, a large fraction of tumors progress to so-called castration-resistant prostate carcinoma (CRPca) which is highly resistant toward chemotherapy. The ensuing high mortality rates illustrate the importance of novel therapeutic targets for CRPCa. The transcription factor NF-κB was recently proposed as such a potential target for therapeutic intervention in CRPCa. Although NF-κB is essential for the regulation of innate and adaptive immunity recent data suggest a role of NF-κB in cancer initiation and progression. However, the exact function of NF-κB signaling in PCa is still a matter of debate. Here, we review known roles of NF-κB signaling in PCa and emphasize the crosstalk of NF-κB and androgen receptor signaling. Finally, we discuss potential therapeutic relevance of blocking NF-κB in PCa.
Collapse
Affiliation(s)
- Garima Jain
- Institute of Pathology, University of Ulm, Albert-Einstein-Allee 23, 89070, Ulm, Germany
| | | | | | | | | |
Collapse
|
23
|
Cavalcante P, Maggi L, Colleoni L, Caldara R, Motta T, Giardina C, Antozzi C, Berrih-Aknin S, Bernasconi P, Mantegazza R. Inflammation and epstein-barr virus infection are common features of myasthenia gravis thymus: possible roles in pathogenesis. Autoimmune Dis 2011; 2011:213092. [PMID: 21961056 PMCID: PMC3180177 DOI: 10.4061/2011/213092] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 06/23/2011] [Indexed: 12/12/2022] Open
Abstract
The thymus plays a major role in myasthenia gravis (MG). Our recent finding of a persistent Epstein-Barr (EBV) virus infection in some MG thymuses, combined with data showing that the thymus is in a proinflammatory state in most patients, supports a viral contribution to the pathogenesis of MG.
Aim of this study was to gain further evidence for intrathymic chronic inflammation and EBV infection in MG patients. Transcriptional profiling by low density array and real-time PCR showed overexpression of genes involved in inflammatory and immune response in MG thymuses. Real-time PCR for EBV genome, latent (EBER1, EBNA1, LMP1) and lytic (BZLF1) transcripts, and immunohistochemistry for LMP1 and BZLF1 proteins confirmed an active intrathymic EBV infection, further supporting the hypothesis that EBV might contribute to onset or perpetuation of the autoimmune response in MG.
Altogether, our results support a role of inflammation and EBV infection as pathogenic features of MG thymus.
Collapse
Affiliation(s)
- Paola Cavalcante
- Department of Neurology IV, Neuromuscular Diseases and Neuroimmunology, Neurological Institute C. Besta Foundation, 20133 Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Cavalcante P, Le Panse R, Berrih-aknin S, Maggi L, Antozzi C, Baggi F, Bernasconi P, Mantegazza R. The thymus in myasthenia gravis: Site of “innate autoimmunity”? Muscle Nerve 2011; 44:467-84. [DOI: 10.1002/mus.22103] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
25
|
Yew TL, Hung YT, Li HY, Chen HW, Chen LL, Tsai KS, Chiou SH, Chao KC, Huang TF, Chen HL, Hung SC. Enhancement of wound healing by human multipotent stromal cell conditioned medium: the paracrine factors and p38 MAPK activation. Cell Transplant 2010; 20:693-706. [PMID: 21176394 DOI: 10.3727/096368910x550198] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Wound healing can be improved by transplanting mesenchymal stem cells (MSCs). In this study, we have demonstrated the benefits of the conditioned medium derived from human MSCs (CM-MSC) in wound healing using an excisional wound model. CM-MSC accelerated wound closure with increased reepithelialization, cell infiltration, granulation formation, and angiogenesis. Notably, CM-MSC enhanced epithelial and endothelial cell migration, suggesting the contribution of increased cell migration to wound healing enhanced by CM-MSC. Cytokine array, ELISA analysis, and quantitative RT-PCR revealed high levels of IL-6 in CM-MSC. Moreover, IL-6 added to the preconditioned medium enhanced both cell migration and wound healing, and antibodies against IL-6 blocked the increase in cell motility and wound closure by CM-MSC. The IL-6 secretory pathway of MSCs was inhibited by SB203580, an inhibitor of p38 MAPK or siRNA against p38 MAPK, suggesting IL-6 secretion by MSCs is mediated through the activation of p38 MAPK. Inactivation of p38 MAPK also reduced the expression and production of IL-8 and CXCL1 by MSCs, both of which were also demonstrated to enhance cell migration and wound closure. Thus, our data suggest MSCs promote wound healing through releasing a repertoire of paracrine factors via activation of p38 MAPK, and the CM-MSC may be applied to enhance wound healing.
Collapse
Affiliation(s)
- Tu-Lai Yew
- Institute of Oral Biology, Department of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Obarzanek-Fojt M, Favre B, Huber M, Ryser S, Moodycliffe A, Wipff PJ, Hinz B, Hohl D. Induction of p38, tumour necrosis factor-α and RANTES by mechanical stretching of keratinocytes expressing mutant keratin 10R156H. Br J Dermatol 2010; 164:125-34. [DOI: 10.1111/j.1365-2133.2010.10013.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Le Panse R, Bismuth J, Cizeron-Clairac G, Weiss JM, Cufi P, Dartevelle P, De Rosbo NK, Berrih-Aknin S. Thymic remodeling associated with hyperplasia in myasthenia gravis. Autoimmunity 2010; 43:401-12. [DOI: 10.3109/08916930903563491] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
28
|
Cheng YC, Liang CM, Chen YP, Tsai IH, Kuo CC, Liang SM. F-spondin plays a critical role in murine neuroblastoma survival by maintaining IL-6 expression. J Neurochem 2009; 110:947-55. [PMID: 19549008 DOI: 10.1111/j.1471-4159.2009.06186.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
F-spondin is associated with the regulation of axonal growth and the development of the nervous system. Its mechanism of action, however, is not clearly understood. In this study, we found that murine neuroblastoma Neuro-2a cells expressed a significant level of IL-6, but only trace amounts of IL-12, tumor necrosis factor alpha and nitric oxide. Knock-down of F-spondin mRNA in murine neuroblastoma NB41A3 and Neuro-2a cells using small interfering RNAs led to decreased IL-6 levels along with lower resistance to serum starvation and cytotoxic amyloid beta(1-42) (Abeta(1-42)) peptide. Restoring decline of F-spondin or IL-6 induced by F-spondin knock-down through adding exogenous F-spondin, IL-6 or over-expressing F-spondin reversed the cell death induced by Abeta(1-42) peptide or serum starvation. The decrease of IL-6 level was positively correlated with decrease of NF-kappaB and inhibition of p38 mitogen-activated protein kinase (MAPK). Over-expressing MEKK, a kinase activator of the p38 MAPK pathway, increased IL-6 production, restored the decrease of p38 induced by F-spondin knock-down, and rescued the cells from death caused by Abeta(1-42) peptide. Taken together, these results suggest that F-spondin may play a critical role in murine neuroblastoma survival under adverse conditions by maintaining IL-6 level via a MEKK/p38 MAPK/NF-kappaB-dependent pathway.
Collapse
Affiliation(s)
- Yung-Chih Cheng
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
29
|
Epithelial cell survival by activating transcription factor 3 (ATF3) in response to chemical ribosome-inactivating stress. Biochem Pharmacol 2009; 77:1105-15. [DOI: 10.1016/j.bcp.2008.11.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Revised: 11/25/2008] [Accepted: 11/26/2008] [Indexed: 12/14/2022]
|
30
|
Colombatti M, Grasso S, Porzia A, Fracasso G, Scupoli MT, Cingarlini S, Poffe O, Naim HY, Heine M, Tridente G, Mainiero F, Ramarli D. The prostate specific membrane antigen regulates the expression of IL-6 and CCL5 in prostate tumour cells by activating the MAPK pathways. PLoS One 2009; 4:e4608. [PMID: 19242540 PMCID: PMC2643478 DOI: 10.1371/journal.pone.0004608] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Accepted: 01/17/2009] [Indexed: 12/23/2022] Open
Abstract
The interleukin-6 (IL-6) and the chemokine CCL5 are implicated in the development and progression of several forms of tumours including that of the prostate. The expression of the prostate specific membrane antigen (PSMA) is augmented in high-grade and metastatic tumors. Observations of the clinical behaviour of prostate tumors suggest that the increased secretion of IL-6 and CCL5 and the higher expression of PSMA may be correlated. We hypothesized that PSMA could be endowed with signalling properties and that its stimulation might impact on the regulation of the gene expression of IL-6 and CCL5. We herein demonstrate that the cross-linking of cell surface PSMA with specific antibodies activates the small GTPases RAS and RAC1 and the MAPKs p38 and ERK1/2 in prostate carcinoma LNCaP cells. As downstream effects of the PSMA-fostered RAS-RAC1-MAPK pathway activation we observed a strong induction of NF-κB activation associated with an increased expression of IL-6 and CCL5 genes. Pharmacological blockade with specific inhibitors revealed that both p38 and ERK1/2 participate in the phenomenon, although a major role exerted by p38 was evident. Finally we demonstrate that IL-6 and CCL5 enhanced the proliferative potential of LNCaP cells synergistically and in a dose-dependent manner and that CCL5 functioned by receptor-mediated activation of the STAT5-Cyclin D1 pro-proliferative pathway. The novel functions attributable to PSMA which are described in the present report may have profound influence on the survival and proliferation of prostate tumor cells, accounting for the observation that PSMA overexpression in prostate cancer patients is related to a worse prognosis.
Collapse
Affiliation(s)
| | - Silvia Grasso
- Department of Pathology, University of Verona, Verona, Italy
| | - Alessandra Porzia
- Department of Experimental Medicine, Institute Pasteur-Fondazione Cenci Bolognetti, University of Rome “La Sapienza”, Rome, Italy
| | - Giulio Fracasso
- Department of Pathology, University of Verona, Verona, Italy
| | - Maria Teresa Scupoli
- Interdepartmental Laboratory for Medical Research (LURM), University of Verona, Verona, Italy
| | - Sara Cingarlini
- Department of Pathology, University of Verona, Verona, Italy
| | - Ornella Poffe
- Department of Pathology, University of Verona, Verona, Italy
| | - Hassan Y. Naim
- Department of Physiological Chemistry, School of Veterinary Medicine, Hannover, Germany
| | - Martin Heine
- Department of Physiological Chemistry, School of Veterinary Medicine, Hannover, Germany
| | - Giuseppe Tridente
- Department of Pathology, University of Verona, Verona, Italy
- Clinical Immunology, Giovanni Battista Rossi Hospital, Verona, Italy
| | - Fabrizio Mainiero
- Department of Experimental Medicine, Institute Pasteur-Fondazione Cenci Bolognetti, University of Rome “La Sapienza”, Rome, Italy
| | - Dunia Ramarli
- Clinical Immunology, Giovanni Battista Rossi Hospital, Verona, Italy
- * E-mail:
| |
Collapse
|
31
|
Sommer N, Tackenberg B, Hohlfeld R. The immunopathogenesis of myasthenia gravis. HANDBOOK OF CLINICAL NEUROLOGY 2008; 91:169-212. [PMID: 18631843 DOI: 10.1016/s0072-9752(07)01505-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Affiliation(s)
- Norbert Sommer
- Clinical Neuroimmunology Group, Philipps-University, Marburg, Germany
| | | | | |
Collapse
|
32
|
Kalamida D, Poulas K, Avramopoulou V, Fostieri E, Lagoumintzis G, Lazaridis K, Sideri A, Zouridakis M, Tzartos SJ. Muscle and neuronal nicotinic acetylcholine receptors. FEBS J 2007; 274:3799-845. [PMID: 17651090 DOI: 10.1111/j.1742-4658.2007.05935.x] [Citation(s) in RCA: 219] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are integral membrane proteins and prototypic members of the ligand-gated ion-channel superfamily, which has precursors in the prokaryotic world. They are formed by the assembly of five transmembrane subunits, selected from a pool of 17 homologous polypeptides (alpha1-10, beta1-4, gamma, delta, and epsilon). There are many nAChR subtypes, each consisting of a specific combination of subunits, which mediate diverse physiological functions. They are widely expressed in the central nervous system, while, in the periphery, they mediate synaptic transmission at the neuromuscular junction and ganglia. nAChRs are also found in non-neuronal/nonmuscle cells (keratinocytes, epithelia, macrophages, etc.). Extensive research has determined the specific function of several nAChR subtypes. nAChRs are now important therapeutic targets for various diseases, including myasthenia gravis, Alzheimer's and Parkinson's diseases, and schizophrenia, as well as for the cessation of smoking. However, knowledge is still incomplete, largely because of a lack of high-resolution X-ray structures for these molecules. Nevertheless, electron microscopy studies on 2D crystals of nAChR from fish electric organs and the determination of the high-resolution X-ray structure of the acetylcholine binding protein (AChBP) from snails, a homolog of the extracellular domain of the nAChR, have been major steps forward and the data obtained have important implications for the design of subtype-specific drugs. Here, we review some of the latest advances in our understanding of nAChRs and their involvement in physiology and pathology.
Collapse
Affiliation(s)
- Dimitra Kalamida
- Department of Pharmacy, University of Patras, Rio Patras, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Fostieri E, Kostelidou K, Poulas K, Tzartos SJ. Recent advances in the understanding and therapy of myasthenia gravis. FUTURE NEUROLOGY 2006. [DOI: 10.2217/14796708.1.6.799] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Myasthenia gravis (MG) is a T-cell dependent autoimmune disease mediated by autoantibodies, which mainly target muscle nicotinic acetylcholine receptors (AChR) and cause loss of functional AChRs in the neuromuscular junction. Both MG and its major autoantigen are studied extensively, yet the etiology of the disease remains unclear, although it is known to be associated with the thymus. A genetic predisposition, combined with several unidentified environmental stimuli, likely creates a favorable milieu in which the disease can appear. Current research focusses on elucidating the cellular and molecular pathways of immune dysregulation, which underly MG outburst and progression. Considerable progress has been made concerning the involvement of the thymus, the identification of impaired mechanisms of immune control and the B–T-cell interaction in MG pathogenesis, while the role of chemokines arises as an intriguing new puzzle. Recent findings fueled the development of novel therapeutic approaches with some encouraging, although preliminary, results. This review summarizes recent achievements in the fields of both basic research and therapeutics.
Collapse
Affiliation(s)
- Efrosini Fostieri
- Hellenic Pasteur Institute, Department of Biochemistry, 127 Vas. Sofias Avenue, 11521 Athens, Greece
| | - Kalliopi Kostelidou
- Hellenic Pasteur Institute, Department of Biochemistry, 127 Vas. Sofias Avenue 11521 Athens, Greece
| | | | - Socrates J Tzartos
- Hellenic Pasteur Institute, Department of Biochemistry, 127 Vas. Sofias Avenue, 11521 Athens, Greece and, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| |
Collapse
|
34
|
Sassano P, Paparo F, Ramieri V, Colangeli W, Verdino G. Interleukine-6 (IL-6) may be a link between myasthenia gravis and myoepithelioma of the parotid gland. Med Hypotheses 2006; 68:314-7. [PMID: 16997495 DOI: 10.1016/j.mehy.2006.06.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Accepted: 06/22/2006] [Indexed: 11/21/2022]
Abstract
Myoepithelioma is a rare benign neoplasm of the salivary glands occurring more frequently in the parotids. Myasthenia gravis (MG) is a chronic, T-cell dependent, antibody and complement-mediated autoimmune neuromuscular transmission disorder. Interleukine-6 (IL-6) is an immune protein belonging to the family of the hematopoietins, liberated in response to infection, burns, trauma, and neoplastic diseases. It seems that an overproduction of IL-6 might play an important role in the pathophysiology of MG. Moreover, it has been discussed the possible role of IL-6 as a modulating factor either in proliferation or in differentiation of pleomorphic adenoma cell line into myoepithelioma. The authors present a rare case of parotid myoepithelioma occurred in a patient affected by myasthenia gravis and suppose a possible IL-6 mediated relationship between myasthenia gravis and parotid myoepithelioma.
Collapse
Affiliation(s)
- Pierpaolo Sassano
- Department of Maxillo-Facial Surgery, University La Sapienza of Rome, Viale del Policlinico, 121, 00161 Roma, RM, Italy
| | | | | | | | | |
Collapse
|
35
|
Thangarajh M, Masterman T, Helgeland L, Rot U, Jonsson MV, Eide GE, Pirskanen R, Hillert J, Jonsson R. The thymus is a source of B-cell-survival factors-APRIL and BAFF-in myasthenia gravis. J Neuroimmunol 2006; 178:161-6. [PMID: 16820216 DOI: 10.1016/j.jneuroim.2006.05.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 05/22/2006] [Accepted: 05/25/2006] [Indexed: 11/24/2022]
Abstract
The accumulation of B cells in the thymus is a common feature of myasthenia gravis (MG). To understand whether factors enhancing B-cell survival are increased in MG, we studied the expression of APRIL, BAFF and three of their receptors in the thymus. In hyperplastic thymi, macrophages expressed APRIL and BAFF, and germinal-center B cells, BAFF-R. CD138-positive plasma cells were abundant in MG thymi. By contrast, BCMA-positive plasma cells were scarce. The expression of APRIL and BAFF in MG thymi may reflect the establishment of an environment favorable to B-cell survival.
Collapse
Affiliation(s)
- Mathula Thangarajh
- Neurology Division, Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|