1
|
Ai X, Yu H, Cai Y, Guan Y. Interactions Between Extracellular Vesicles and Autophagy in Neuroimmune Disorders. Neurosci Bull 2024; 40:992-1006. [PMID: 38421513 PMCID: PMC11251008 DOI: 10.1007/s12264-024-01183-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 11/15/2023] [Indexed: 03/02/2024] Open
Abstract
Neuroimmune disorders, such as multiple sclerosis, neuromyelitis optica spectrum disorder, myasthenia gravis, and Guillain-Barré syndrome, are characterized by the dysfunction of both the immune system and the nervous system. Increasing evidence suggests that extracellular vesicles and autophagy are closely associated with the pathogenesis of these disorders. In this review, we summarize the current understanding of the interactions between extracellular vesicles and autophagy in neuroimmune disorders and discuss their potential diagnostic and therapeutic applications. Here we highlight the need for further research to fully understand the mechanisms underlying these disorders, and to develop new diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Xiwen Ai
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200127, China
| | - Haojun Yu
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200127, China
| | - Yu Cai
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
| | - Yangtai Guan
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200127, China.
| |
Collapse
|
2
|
Huang EJC, Wu MH, Wang TJ, Huang TJ, Li YR, Lee CY. Myasthenia Gravis: Novel Findings and Perspectives on Traditional to Regenerative Therapeutic Interventions. Aging Dis 2023; 14:1070-1092. [PMID: 37163445 PMCID: PMC10389825 DOI: 10.14336/ad.2022.1215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/15/2022] [Indexed: 05/12/2023] Open
Abstract
The prevalence of myasthenia gravis (MG), an autoimmune disorder, is increasing among all subsets of the population leading to an elevated economic and social burden. The pathogenesis of MG is characterized by the synthesis of autoantibodies against the acetylcholine receptor (AChR), low-density lipoprotein receptor-related protein 4 (LRP4), or muscle-specific kinase at the neuromuscular junction, thereby leading to muscular weakness and fatigue. Based on clinical and laboratory examinations, the research is focused on distinguishing MG from other autoimmune, genetic diseases of neuromuscular transmission. Technological advancements in machine learning, a subset of artificial intelligence (AI) have been assistive in accurate diagnosis and management. Besides, addressing the clinical needs of MG patients is critical to improving quality of life (QoL) and satisfaction. Lifestyle changes including physical exercise and traditional Chinese medicine/herbs have also been shown to exert an ameliorative impact on MG progression. To achieve enhanced therapeutic efficacy, cholinesterase inhibitors, immunosuppressive drugs, and steroids in addition to plasma exchange therapy are widely recommended. Under surgical intervention, thymectomy is the only feasible alternative to removing thymoma to overcome thymoma-associated MG. Although these conventional and current therapeutic approaches are effective, the associated adverse events and surgical complexity limit their wide application. Moreover, Restivo et al. also, to increase survival and QoL, further recent developments revealed that antibody, gene, and regenerative therapies (such as stem cells and exosomes) are currently being investigated as a safer and more efficacious alternative. Considering these above-mentioned points, we have comprehensively reviewed the recent advances in pathological etiologies of MG including COVID-19, and its therapeutic management.
Collapse
Affiliation(s)
- Evelyn Jou-Chen Huang
- Department of Ophthalmology, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Meng-Huang Wu
- Department of Orthopedics, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Tsung-Jen Wang
- Department of Ophthalmology, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Tsung-Jen Huang
- Department of Orthopedics, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Yan-Rong Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Ching-Yu Lee
- Department of Orthopedics, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
3
|
Huang H, Ran H, Liu X, Yu L, Qiu L, Lin Z, Ou C, Lu Y, Yang W, Liu W. Leflunomide ameliorates experimental autoimmune myasthenia gravis by regulating humoral and cellular immune responses. Int Immunopharmacol 2021; 93:107434. [PMID: 33556668 DOI: 10.1016/j.intimp.2021.107434] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/12/2022]
Abstract
Leflunomide, an immunosuppressive disease-modifying anti-rheumatic drug (DMARD), is widely used in the treatment of rheumatoid arthritis (RA), psoriatic arthritis (PA) as well as multiple sclerosis. However, its role in myasthenia gravis (MG) has not yet been clearly explored. Here, we investigated the effect of leflunomide on experimental autoimmune myasthenia gravis (EAMG) in vivo and in vitro. The results demonstrated that leflunomide alleviated the severity of EAMG associated with reduced serum total anti-acetylcholine receptor (AChR) IgG levels. During the development of EAMG, the increase of follicular helper T cells (Tfh) 1, Tfh 17 cells and decrease of follicular regulatory T cells (Tfr) were reversely altered after leflunomide administration. Our work further found that leflunomide might inhibit Tfh cells through the IL-21/STAT3 pathway to reduce the secretion of antibodies by B cells. In addition, leflunomide rebuilt the balance of Th1/Th2/Th17/Treg subsets. These results suggested that leflunomide ameliorated EAMG severity by regulating humoral immune responses and Th cell profiles thereby providing a novel effective treatment strategy for MG.
Collapse
Affiliation(s)
- Huan Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Hao Ran
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xiaoxi Liu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lu Yu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Li Qiu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zhongqiang Lin
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Changyi Ou
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yaru Lu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Wenhao Yang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Weibin Liu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
4
|
Abstract
Acquired Myasthenia Gravis (MG) is a neuromuscular disease caused by autoantibodies against components of the neuromuscular junction. It is a prototype organ-specific autoimmune disease with well-defined antigenic targets mainly the nicotinic acetylcholine receptor (AChR). Patients suffer from fluctuating, fatigable muscle weakness that worsens with activity and improves with rest. Various therapeutic strategies have been used over the years to alleviate MG symptoms. These strategies aim at improving the transmission of the nerve impulse to muscle or at lowering the immune system with steroids or immunosuppressant drugs. Nevertheless, MG remains a chronic disease and symptoms tend to persist in many patients, some being or becoming refractory over time. In this review, based on recent experimental data on MG or based on results from clinical trials for other autoimmune diseases, we explore new potential therapeutic approaches for MG patients, going from non-specific approaches with the use of stem cells with their anti-inflammatory and immunosuppressive properties to targeted therapies using monoclonal antibodies specific for cell-surface antigens or circulating molecules.
Collapse
Affiliation(s)
- Anthony Behin
- APHP, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France, Institut de Myologie, GH Pitié-Salpêtrière, Paris, France.,AIM, Institut de Myologie, Paris, France
| | - Rozen Le Panse
- INSERM U974, Paris, France.,UPMC Sorbonne Université, Paris, France.,AIM, Institut de Myologie, Paris, France
| |
Collapse
|
5
|
Zheng WP, Zhang BY, Shen ZY, Yin ML, Cao Y, Song HL. Biological effects of bone marrow mesenchymal stem cells on hepatitis B virus in vitro. Mol Med Rep 2017; 15:2551-2559. [PMID: 28447750 PMCID: PMC5428401 DOI: 10.3892/mmr.2017.6330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 12/09/2016] [Indexed: 02/07/2023] Open
Abstract
The aim of the present study was to explore the effects of co‑culturing bone marrow‑derived mesenchymal stem cells (BM-MSCs) cultured with hepatitis B virus (HBV)‑infected lymphocytes in vitro. BM‑MSCs and lymphocytes from Brown Norway rats were obtained from the bone marrow and spleen, respectively. Rats were divided into the following five experimental groups: Group 1, splenic lymphocytes (SLCs); group 2, HepG2.2.15 cells; group 3, BM‑MSCs + HepG2.2.15 cells; group 4, SLCs + HepG2.2.15 cells; and group 5, SLCs + BM‑MSCs + HepG2.2.15 cells. The viability of lymphocytes and HepG2.2.15 cells was assessed using the MTT assay at 24, 48 and 72 h, respectively. Levels of supernatant HBV DNA and intracellular HBV covalently closed circular DNA (cccDNA) were measured using quantitative polymerase chain reaction. Supernatant cytokine levels were measured by enzyme‑linked immunosorbent assay (ELISA). T cell subsets were quantified by flow cytometry using fluorescence‑labeled antibodies. In addition, the HBV genome sequence was analyzed by direct gene sequencing. Levels of HBV DNA and cccDNA in group 5 were lower when compared with those in group 3 or group 4, with a significant difference observed at 48 h. The secretion of interferon‑γ was negatively correlated with the level of HBV DNA, whereas secretion of interleukin (IL)‑10 and IL‑22 were positively correlated with the level of HBV DNA. Flow cytometry demonstrated that the percentage of CD3+CD8+ T cells was positively correlated with the levels of HBV DNA, and the CD3+CD4+/CD3+CD8+ ratio was negatively correlated with the level of HBV DNA. Almost no mutations in the HBV DNA sequence were detected in HepG2.2.15 cells co‑cultured with BM‑MSCs, SLCs, or in the two types of cells combined. BM‑MSCs inhibited the expression of HBV DNA and enhanced the clearance of HBV, which may have been mediated by the regulation of the Tc1/Tc2 cell balance and the mode of cytokine secretion to modulate cytokine expression.
Collapse
Affiliation(s)
- Wei-Ping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Bo-Ya Zhang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Zhong-Yang Shen
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Ming-Li Yin
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yi Cao
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Hong-Li Song
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
- Tianjin Key Laboratory of Organ Transplantation, Tianjin 300192, P.R. China
| |
Collapse
|
6
|
MM-BMSCs induce naïve CD4+ T lymphocytes dysfunction through fibroblast activation protein α. Oncotarget 2017; 8:52614-52628. [PMID: 28881756 PMCID: PMC5581055 DOI: 10.18632/oncotarget.17538] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/11/2017] [Indexed: 01/14/2023] Open
Abstract
Background The tumor microenvironment plays a major role in multiple myelomas (MM). MM-BMSCs (bone marrow mesenchymal stromal cells) can support tumor growth and immune surveillance escape. On the other hand, fibroblast activation protein α, expressed by cancer stroma cells including BMSCs, has been shown to potentiate epithelial cancers growth and immune suppression. Results MM-BMSC inhibited proliferation of T cells (P = 0.0138), promoted senescence of T cells (P < 0.001), consistent with decreased CD28 and hTERT expression (P < 0.001), Treg/Th17 was down-regulated by MM-BMSC (P = 0.031). After treatment with FAPα inhibitor PT-100, senescent rate was decreased (P = 0.001), Treg/Th17 was up-regulated (P = 0.024). FAPα was up-regulated by TCCM (P = 0.02). p-AKT was increased in MM-BMSC co-cultured T cells (P = 0.021) and decreased by PT-100 (P = 0.017). Higher level of TGF-β was observed in MM-BMSC co-cultured medium (P < 0.001), and down-regulated by PT-100 (P = 0.038). p-AKT was upregulated as compared to T-cells without MM-BMSCs (P = 0.021). The abnormal p-AKT level was distinctly decreased by PT-100 (P = 0.017). Materials and Methods The expression of FAPα was analyzed by western blot and RT-PCR. The proliferation and senescence of CD4+ T cells was examined by cck-8 and β-gal staining, and Treg/Th17, CD28 expression was analyzed by FCM. The FAPα and PI3K pathway was analyzed by western blot and their relationship with T cell function was detected by FCM and RT-PCR. The level of IL-10, IL-17 and TGF-β was detected by ELISA. Conclusions MM-BMSCs inhibit T-cell proliferation and drive Th17 differentiation through FAPα/TGF-β axis, leading to the progression of myeloma. FAPα-induced T-cell senescence is mediated by the PI3K signaling pathway.
Collapse
|
7
|
Sudres M, Maurer M, Robinet M, Bismuth J, Truffault F, Girard D, Dragin N, Attia M, Fadel E, Santelmo N, Sicsic C, Brenner T, Berrih-Aknin S. Preconditioned mesenchymal stem cells treat myasthenia gravis in a humanized preclinical model. JCI Insight 2017; 2:e89665. [PMID: 28405609 DOI: 10.1172/jci.insight.89665] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Myasthenia gravis (MG) with anti-acetylcholine receptor (AChR) Abs is an autoimmune disease characterized by severe defects in immune regulation and thymic inflammation. Because mesenchymal stem cells (MSCs) display immunomodulatory features, we investigated whether and how in vitro-preconditioned human MSCs (cMSCs) could treat MG disease. We developed a new humanized preclinical model by subcutaneously grafting thymic MG fragments into immunodeficient NSG mice (NSG-MG model). Ninety percent of the animals displayed human anti-AChR Abs in the serum, and 50% of the animals displayed MG-like symptoms that correlated with the loss of AChR at the muscle endplates. Interestingly, each mouse experiment recapitulated the MG features of each patient. We next demonstrated that cMSCs markedly improved MG, reducing the level of anti-AChR Abs in the serum and restoring AChR expression at the muscle endplate. Resting MSCs had a smaller effect. Finally, we showed that the underlying mechanisms involved (a) the inhibition of cell proliferation, (b) the inhibition of B cell-related and costimulatory molecules, and (c) the activation of the complement regulator DAF/CD55. In conclusion, this study shows that a preconditioning step promotes the therapeutic effects of MSCs via combined mechanisms, making cMSCs a promising strategy for treating MG and potentially other autoimmune diseases.
Collapse
Affiliation(s)
- Muriel Sudres
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Marie Maurer
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Marieke Robinet
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Jacky Bismuth
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Frédérique Truffault
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Diane Girard
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Nadine Dragin
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Mohamed Attia
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Elie Fadel
- Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | | | - Camille Sicsic
- Department of Neurology, Agnes Ginges Center for human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Talma Brenner
- Department of Neurology, Agnes Ginges Center for human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Sonia Berrih-Aknin
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| |
Collapse
|
8
|
Intravenous Administration of Adipose-Derived Stem Cell Protein Extracts Improves Neurological Deficits in a Rat Model of Stroke. Stem Cells Int 2017; 2017:2153629. [PMID: 28265288 PMCID: PMC5318632 DOI: 10.1155/2017/2153629] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/21/2016] [Accepted: 11/27/2016] [Indexed: 12/14/2022] Open
Abstract
Treatment of adipose-derived stem cell (ADSC) substantially improves the neurological deficits during stroke by reducing neuronal injury, limiting proinflammatory immune responses, and promoting neuronal repair, which makes ADSC-based therapy an attractive approach for treating stroke. However, the potential risk of tumorigenicity and low survival rate of the implanted cells limit the clinical use of ADSC. Cell-free extracts from ADSC (ADSC-E) may be a feasible approach that could overcome these limitations. Here, we aim to explore the potential usage of ADSC-E in treating rat transient middle cerebral artery occlusion (tMCAO). We demonstrated that intravenous (IV) injection of ADSC-E remarkably reduces the ischemic lesion and number of apoptotic neurons as compared to other control groups. Although ADSC and ADSC-E treatment results in a similar degree of a long-term clinical beneficial outcome, the dynamics between two ADSC-based therapies are different. While the injection of ADSC leads to a relatively mild but prolonged therapeutic effect, the administration of ADSC-E results in a fast and pronounced clinical improvement which was associated with a unique change in the molecular signature suggesting that potential mechanisms underlying different therapeutic approach may be different. Together these data provide translational evidence for using protein extracts from ADSC for treating stroke.
Collapse
|
9
|
Maria ATJ, Maumus M, Le Quellec A, Jorgensen C, Noël D, Guilpain P. Adipose-Derived Mesenchymal Stem Cells in Autoimmune Disorders: State of the Art and Perspectives for Systemic Sclerosis. Clin Rev Allergy Immunol 2016; 52:234-259. [DOI: 10.1007/s12016-016-8552-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
10
|
Indoleamine 2,3-dioxygenase mediates inhibition of virus-specific CD8+ T cell proliferation by human mesenchymal stromal cells. Cytotherapy 2016; 18:621-9. [DOI: 10.1016/j.jcyt.2016.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 01/21/2016] [Accepted: 01/22/2016] [Indexed: 01/08/2023]
|
11
|
Mantegazza R, Cordiglieri C, Consonni A, Baggi F. Animal models of myasthenia gravis: utility and limitations. Int J Gen Med 2016; 9:53-64. [PMID: 27019601 PMCID: PMC4786081 DOI: 10.2147/ijgm.s88552] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Myasthenia gravis (MG) is a chronic autoimmune disease caused by the immune attack of the neuromuscular junction. Antibodies directed against the acetylcholine receptor (AChR) induce receptor degradation, complement cascade activation, and postsynaptic membrane destruction, resulting in functional reduction in AChR availability. Besides anti-AChR antibodies, other autoantibodies are known to play pathogenic roles in MG. The experimental autoimmune MG (EAMG) models have been of great help over the years in understanding the pathophysiological role of specific autoantibodies and T helper lymphocytes and in suggesting new therapies for prevention and modulation of the ongoing disease. EAMG can be induced in mice and rats of susceptible strains that show clinical symptoms mimicking the human disease. EAMG models are helpful for studying both the muscle and the immune compartments to evaluate new treatment perspectives. In this review, we concentrate on recent findings on EAMG models, focusing on their utility and limitations.
Collapse
Affiliation(s)
- Renato Mantegazza
- Neurology IV Unit, Neuroimmunology and Neuromuscular Disorders, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy
| | - Chiara Cordiglieri
- Neurology IV Unit, Neuroimmunology and Neuromuscular Disorders, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy
| | - Alessandra Consonni
- Neurology IV Unit, Neuroimmunology and Neuromuscular Disorders, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy
| | - Fulvio Baggi
- Neurology IV Unit, Neuroimmunology and Neuromuscular Disorders, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy
| |
Collapse
|
12
|
Wang X, Zhao W, Wang J, Shi K, Qin X, Kong Q, Wang G, Mu L, Li H, Sun B, Shi L. Bone Marrow Stromal Cells Inhibit the Activation of Liver Cirrhotic Fat-Storing Cells via Adrenomedullin Secretion. Dig Dis Sci 2015; 60:1325-34. [PMID: 25445161 DOI: 10.1007/s10620-014-3423-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 10/30/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cirrhosis, or liver fibrosis, which is mainly triggered by cirrhosis fat-storing cells (CFSCs) activation, has traditionally been considered an irreversible disease. However, recent observations indicate that even advanced fibrosis is still reversible by removing the causative agents. Anti-fibrotic effects of bone marrow-derived stromal cells (BMSCs) have been demonstrated by inhibiting CFSCs via cytokines secretion; however, the mechanisms are still unclear. AIMS The purpose of this study was to explore the underlying mechanisms by which BMSCs modulate the function of activated CFSCs. METHODS After the co-culture of CFSCs with BMSCs supernatants with or without the addition of recombinant rat adrenomedullin (AM)/AM-specific siRNA, western blot analysis was mainly used to detect the differences of relative protein expression on CFSCs. RESULTS BMSC-secreted adrenomedullin (AM) effectively inhibited the proliferation and activation of CFSCs by suppressing the expression of Ang II and its binding receptor, AT1, which resulted in a reduction of p47-phox formation. CONCLUSIONS Our data suggested that BMSCs inhibited CFSC activation in vitro via the AM-Ang II-p47-phox signaling pathway, and since CFSC activation is an essential part of hepatic fibrosis process, this inhibition by BMSCs implies us new insights into the potential treatment of hepatic fibrosis via BMSCs.
Collapse
Affiliation(s)
- Xiaodong Wang
- Emergency Department, The 2nd Affiliated Hospital of Harbin Medical University, 148 Bao Jian Road, Nangang District, Harbin, 150086, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Losen M, Martinez-Martinez P, Molenaar PC, Lazaridis K, Tzartos S, Brenner T, Duan RS, Luo J, Lindstrom J, Kusner L. Standardization of the experimental autoimmune myasthenia gravis (EAMG) model by immunization of rats with Torpedo californica acetylcholine receptors--Recommendations for methods and experimental designs. Exp Neurol 2015; 270:18-28. [PMID: 25796590 PMCID: PMC4466156 DOI: 10.1016/j.expneurol.2015.03.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/06/2015] [Accepted: 03/10/2015] [Indexed: 12/21/2022]
Abstract
Myasthenia gravis (MG) with antibodies against the acetylcholine receptor (AChR) is characterized by a chronic, fatigable weakness of voluntary muscles. The production of autoantibodies involves the dysregulation of T cells which provide the environment for the development of autoreactive B cells. The symptoms are caused by destruction of the postsynaptic membrane and degradation of the AChR by IgG autoantibodies, predominantly of the G1 and G3 subclasses. Active immunization of animals with AChR from mammalian muscles, AChR from Torpedo or Electrophorus electric organs, and recombinant or synthetic AChR fragments generates a chronic model of MG, termed experimental autoimmune myasthenia gravis (EAMG). This model covers cellular mechanisms involved in the immune response against the AChR, e.g. antigen presentation, T cell-help and regulation, B cell selection and differentiation into plasma cells. Our aim is to define standard operation procedures and recommendations for the rat EAMG model using purified AChR from the Torpedo californica electric organ, in order to facilitate more rapid translation of preclinical proof of concept or efficacy studies into clinical trials and, ultimately, clinical practice.
Collapse
Affiliation(s)
- Mario Losen
- Division Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.
| | - Pilar Martinez-Martinez
- Division Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Peter C Molenaar
- Division Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | | | - Socrates Tzartos
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Talma Brenner
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, PR China
| | - Jie Luo
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, USA
| | - Jon Lindstrom
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, USA
| | - Linda Kusner
- Department of Pharmacology & Physiology, The George Washington University, Washington, DC, USA
| |
Collapse
|
14
|
Yao X, Kong Q, Xie X, Wang J, Li N, Liu Y, Sun B, Li Y, Wang G, Li W, Qu S, Zhao H, Wang D, Liu X, Zhang Y, Mu L, Li H. Neutralization of interleukin-9 ameliorates symptoms of experimental autoimmune myasthenia gravis in rats by decreasing effector T cells and altering humoral responses. Immunology 2014; 143:396-405. [PMID: 24850614 DOI: 10.1111/imm.12322] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 05/01/2014] [Accepted: 05/08/2014] [Indexed: 12/21/2022] Open
Abstract
Interleukin-9 (IL-9) was initially thought to be a type 2 T helper (Th2)-associated cytokine involved in the regulation of autoimmune responses by affecting multiple cell types. However, it was recently shown that IL-9-producing CD4+ T cells represent a discrete subset of Th cells, designated Th9 cells. Although Th9 cells have been shown to be important in many diseases, their roles in myasthenia gravis (MG) are unclear. The aim of this study was to determine whether IL-9 and Th9 cells promote the progression of experimental autoimmune myasthenia gravis (EAMG). The results showed that the percentage of Th9 cells changed during the progression of EAMG, accompanied by an up-regulation of IL-9. Blocking IL-9 activity with antibodies against IL-9 inhibited EAMG-associated pathology in rats and reduced serum anti-acetylcholine receptor IgG levels. Neutralization of IL-9 altered the Th subset distribution in EAMG, reducing the number of Th1 cells and increasing the number of regulatory T cells. Administration of an anti-IL-9 antibody may represent an effective therapeutic strategy for MG-associated pathologies or other T-cell- or B-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Xiuhua Yao
- Department of Neurobiology, Provincial Key Laboratory of Neurobiology, Harbin Medical University, Heilongjiang, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Lui PPY, Kong SK, Lau PM, Wong YM, Lee YW, Tan C, Wong OT. Allogeneic tendon-derived stem cells promote tendon healing and suppress immunoreactions in hosts: in vivo model. Tissue Eng Part A 2014; 20:2998-3009. [PMID: 24798058 DOI: 10.1089/ten.tea.2013.0713] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The medium- to long-term healing effect and infiltration of inflammatory cells, after transplantation of allogeneic tendon-derived stem cell (TDSC) to the rat patellar tendon window wound, were examined. Allogeneic patellar TDSCs derived from a green fluorescent protein rat were used. The outcome of tendon healing and the infiltration of inflammatory cells were examined by histology and immunohistochemistry up to week 16 postinjury. The fate of the transplanted cells was examined by ex vivo fluorescent imaging and immunohistochemistry. Our results showed that the transplantation of allogeneic TDSCs promoted tendon healing with no increased risk of ectopic chondro-ossification up to week 16. A low infiltration of T cells, ED1 macrophages, ED2 macrophages, and mast cells in the window wound was obtained. The transplanted TDSCs were found in the window wound at week 1 and 2, but were absent after week 4 postinjury. In conclusion, allogeneic TDSCs promoted tendon repair in the medium to long term and exhibited weak immunoreactions and anti-inflammatory effects in the hosts after transplantation in a rat model. There was no increased risk of ectopic chondro-ossification after TDSC transplantation. The decrease in the number of transplanted cells with time suggested that allogeneic TDSCs did not promote tendon repair through direct differentiation.
Collapse
|
16
|
Kilpinen L, Impola U, Sankkila L, Ritamo I, Aatonen M, Kilpinen S, Tuimala J, Valmu L, Levijoki J, Finckenberg P, Siljander P, Kankuri E, Mervaala E, Laitinen S. Extracellular membrane vesicles from umbilical cord blood-derived MSC protect against ischemic acute kidney injury, a feature that is lost after inflammatory conditioning. J Extracell Vesicles 2013; 2:21927. [PMID: 24349659 PMCID: PMC3860334 DOI: 10.3402/jev.v2i0.21927] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/31/2013] [Accepted: 11/12/2013] [Indexed: 12/21/2022] Open
Abstract
Background Mesenchymal stromal cells (MSC) are shown to have a great therapeutic potential in many immunological disorders. Currently the therapeutic effect of MSCs is considered to be mediated via paracrine interactions with immune cells. Umbilical cord blood is an attractive but still less studied source of MSCs. We investigated the production of extracellular membrane vesicles (MVs) from human umbilical cord blood derived MSCs (hUCBMSC) in the presence (MVstim) or absence (MVctrl) of inflammatory stimulus. Methods hUCBMSCs were cultured in serum free media with or without IFN-γ and MVs were collected from conditioned media by ultracentrifugation. The protein content of MVs were analyzed by mass spectrometry. Hypoxia induced acute kidney injury rat model was used to analyze the in vivo therapeutic potential of MVs and T-cell proliferation and induction of regulatory T cells were analyzed by co-culture assays. Results Both MVstim and MVctrl showed similar T-cell modulation activity in vitro, but only MVctrls were able to protect rat kidneys from reperfusion injury in vivo. To clarify this difference in functionality we made a comparative mass spectrometric analysis of the MV protein contents. The IFN-γ stimulation induced dramatic changes in the protein content of the MVs. Complement factors (C3, C4A, C5) and lipid binding proteins (i.e apolipoproteins) were only found in the MVctrls, whereas the MVstim contained tetraspanins (CD9, CD63, CD81) and more complete proteasome complex accompanied with MHCI. We further discovered that differently produced MV pools contained specific Rab proteins suggesting that same cells, depending on external signals, produce vesicles originating from different intracellular locations. Conclusions We demonstrate by both in vitro and in vivo models accompanied with a detailed analysis of molecular characteristics that inflammatory conditioning of MSCs influence on the protein content and functional properties of MVs revealing the complexity of the MSC paracrine regulation.
Collapse
Affiliation(s)
| | - Ulla Impola
- Finnish Red Cross Blood Service, Helsinki, Finland
| | | | - Ilja Ritamo
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - Maria Aatonen
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | | | | | - Leena Valmu
- Finnish Red Cross Blood Service, Helsinki, Finland
| | | | - Piet Finckenberg
- Department of Pharmacology, Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Pia Siljander
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland ; Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | | | | |
Collapse
|
17
|
Wang J, Zheng S, Xin N, Dou C, Fu L, Zhang X, Chen J, Zhang Y, Geng D, Xiao C, Cui G, Shen X, Lu Y, Wang J, Dong R, Qiao Y, Zhang Y. Identification of Novel MicroRNA Signatures Linked to Experimental Autoimmune Myasthenia Gravis Pathogenesis: Down-Regulated miR-145 Promotes Pathogenetic Th17 Cell Response. J Neuroimmune Pharmacol 2013; 8:1287-302. [DOI: 10.1007/s11481-013-9498-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 08/26/2013] [Indexed: 12/21/2022]
|
18
|
|
19
|
Abstract
PURPOSE OF REVIEW Mesenchymal stromal cells (MSCs) possess unique immunomodulatory features. MSCs dampen effector T-cell response while promoting the emergence of regulatory T cells. By skewing this balance, MSC could represent the ideal strategy for tolerance induction in organ transplantation. Here we review recent evidence on the efficacy of MSC-based therapy in experimental models of solid organ transplantation as well as the early clinical experiences in kidney transplantation. RECENT FINDINGS MSC infusion in experimental models of solid organ transplantation resulted in a Treg-mediated tolerance. MSC also synergized with low-dose or transient pharmacological immunosuppression in inducing long-term graft survival indicating that these cells could allow safe minimization of maintenance drug therapy. Early results from clinical studies in kidney transplant recipients reported encouraging results on the immunoregulatory effect of MSC, although posttransplant MSC infusion could associate with acute graft dysfunction (engraftment syndrome). SUMMARY Immunoregulatory functions of MSC are not fixed but rather the result of microenvironment they encounter in vivo. Further studies are needed to establish how and wherein these cells have to be administered and how they may function to safely modulate host immune response in vivo in clinical transplant setting.
Collapse
|
20
|
Ma S, Zhong D, Chen H, Zheng Y, Sun Y, Luo J, Li H, Li G, Yin Y. The immunomodulatory effect of bone marrow stromal cells (BMSCs) on interleukin (IL)-23/IL-17-mediated ischemic stroke in mice. J Neuroimmunol 2013; 257:28-35. [DOI: 10.1016/j.jneuroim.2013.01.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 01/15/2013] [Accepted: 01/20/2013] [Indexed: 02/01/2023]
|
21
|
Díaz-Manera J, Rojas García R, Illa I. Treatment strategies for myasthenia gravis: an update. Expert Opin Pharmacother 2012; 13:1873-83. [DOI: 10.1517/14656566.2012.705831] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
22
|
Li N, Mu L, Wang J, Zhang J, Xie X, Kong Q, Tang W, Yao X, Liu Y, Wang L, Wang G, Wang D, Jin L, Sun B, Li H. Activation of the adenosine A2A receptor attenuates experimental autoimmune myasthenia gravis severity. Eur J Immunol 2012; 42:1140-51. [PMID: 22539289 DOI: 10.1002/eji.201142088] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The adenosine A2A receptor (A2AR) is the major cellular adenosine receptor commonly associated with immunosuppression. Here, we investigated whether A2AR activation holds the potential for impacting the severity of experimental autoimmune myasthenia gravis (EAMG) induced following immunization of Lewis rats with the acetylcholine receptor (AChR) R97-116 peptide. This report demonstrates reduced A2AR expression by both T cells and B cells residing in spleen and lymph nodes following EAMG induction. A2AR stimulation inhibited anti-AChR antibody production and proliferation of AChR-specific lymphocytes in vitro. Inhibition was blocked with the A2AR antagonists or protein kinase A inhibitor. We also determined that the development of EAMG was accompanied by a T-helper cell imbalance that could be restored following A2AR stimulation that resulted in increased Treg cell levels and a reduction in Th1-, Th2-, and Th17-cell subtypes. An EAMG-preventive treatment regimen was established that consisted of (2-(p-(2-carbonylethyl)phenylethylamino)-5-N-ethylcarboxamidoadenosine) (CGS21680; A2AR agonist) administration 1 day prior to EAMG induction. Administration of CGS21680 29 days post EAMG induction (therapeutic treatment) also ameliorated disease severity. We conclude that A2AR agonists may represent a new class of compounds that can be developed for use in the treatment of myasthenia gravis or other T-cell- and B-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Na Li
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Baggi F, Antozzi C, Toscani C, Cordiglieri C. Acetylcholine Receptor-Induced Experimental Myasthenia Gravis: What Have We Learned from Animal Models After Three Decades? Arch Immunol Ther Exp (Warsz) 2011; 60:19-30. [DOI: 10.1007/s00005-011-0158-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 09/28/2011] [Indexed: 01/23/2023]
|
24
|
Abstract
Bone marrow is thought to be a primary hematopoietic organ. However, accumulated evidences demonstrate that active function and trafficking of immune cells, including regulatory T cells, conventional T cells, B cells, dendritic cells, natural killer T (NKT) cells, neutrophils, myeloid-derived suppressor cells and mesenchymal stem cells, are observed in the bone marrow. Furthermore, bone marrow is a predetermined metastatic location for multiple human tumors. In this review, we discuss the immune network in the bone marrow. We suggest that bone marrow is an immune regulatory organ capable of fine tuning immunity and may be a potential therapeutic target for immunotherapy and immune vaccination.
Collapse
|
25
|
Duffy MM, Ritter T, Ceredig R, Griffin MD. Mesenchymal stem cell effects on T-cell effector pathways. Stem Cell Res Ther 2011; 2:34. [PMID: 21861858 PMCID: PMC3219065 DOI: 10.1186/scrt75] [Citation(s) in RCA: 334] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem (stromal) cells (MSCs) are rare, multipotent progenitor cells that can be isolated and expanded from bone marrow and other tissues. Strikingly, MSCs modulate the functions of immune cells, including T cells, B cells, natural killer cells, monocyte/macrophages, dendritic cells, and neutrophils. T cells, activated to perform a range of different effector functions, are the primary mediators of many autoimmune and inflammatory diseases as well as of transplant rejection and graft-versus-host disease. Well-defined T-cell effector phenotypes include the CD4+ (T helper cell) subsets Th1, Th2, and Th17 cells and cytotoxic T lymphocytes derived from antigen-specific activation of naïve CD8+ precursors. In addition, naturally occurring and induced regulatory T cells (Treg) represent CD4+ and CD8+ T-cell phenotypes that potently suppress effector T cells to prevent autoimmunity, maintain self-tolerance, and limit inflammatory tissue injury. Many immune-mediated diseases entail an imbalance between Treg and effector T cells of one or more phenotypes. MSCs broadly suppress T-cell activation and proliferation in vitro via a plethora of soluble and cell contact-dependent mediators. These mediators may act directly upon T cells or indirectly via modulation of antigen-presenting cells and other accessory cells. MSC administration has also been shown to be variably associated with beneficial effects in autoimmune and transplant models as well as in several human clinical trials. In a small number of studies, however, MSC administration has been found to aggravate T cell-mediated tissue injury. The multiple effects of MSCs on cellular immunity may reflect their diverse influences on the different T-cell effector subpopulations and their capacity to specifically protect or induce Treg populations. In this review, we focus on findings from the recent literature in which specific modulatory effects of MSCs on one or more individual effector T-cell subsets and Treg phenotypes have been examined in vitro, in relevant animal models of in vivo immunological disease, and in human subjects. We conclude that MSCs have the potential to directly or indirectly inhibit disease-associated Th1, Th2, and Th17 cells as well as cytotoxic T lymphocytes but that many key questions regarding the potency, specificity, mechanistic basis, and predictable therapeutic value of these modulatory effects remain unanswered.
Collapse
Affiliation(s)
- Michelle M Duffy
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and School of Medicine, Nursing and Health Sciences, Orbsen Building, National University of Ireland Galway, Ireland
| | | | | | | |
Collapse
|
26
|
Tüzün E, Huda R, Christadoss P. Complement and cytokine based therapeutic strategies in myasthenia gravis. J Autoimmun 2011; 37:136-43. [PMID: 21636248 DOI: 10.1016/j.jaut.2011.05.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 05/02/2011] [Indexed: 01/17/2023]
Abstract
Myasthenia gravis (MG) is a T cell-dependent and antibody-mediated disease in which the target antigen is the skeletal muscle acetylcholine receptor (AChR). In the last few decades, several immunological factors involved in MG pathogenesis have been discovered mostly by studies utilizing the experimental autoimmune myasthenia gravis (EAMG) model. Nevertheless, MG patients are still treated with non-specific global immunosuppression that is associated with severe chronic side effects. Due to the high heterogeneity of AChR epitopes and antibody responses involved in MG pathogenesis, the specific treatment of MG symptoms have to be achieved by inhibiting the complement factors and cytokines involved in anti-AChR immunity. EAMG studies have clearly shown that inhibition of the classical and common complement pathways effectively and specifically diminish the neuromuscular junction destruction induced by anti-AChR antibodies. The inborn or acquired deficiencies of IL-6, TNF-α and TNF receptor functions are associated with the lowest EAMG incidences. Th17-type immunity has recently emerged as an important contributor of EAMG pathogenesis. Overall, these results suggest that inhibition of the complement cascade and the cytokine networks alone or in combination might aid in development of future treatment models that would reduce MG symptoms with highest efficacy and lowest side effect profile.
Collapse
Affiliation(s)
- Erdem Tüzün
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| | | | | |
Collapse
|
27
|
Wu Y, Ren M, Yang R, Liang X, Ma Y, Tang Y, Huang L, Ye J, Chen K, Wang P, Shen H. Reduced immunomodulation potential of bone marrow-derived mesenchymal stem cells induced CCR4+CCR6+ Th/Treg cell subset imbalance in ankylosing spondylitis. Arthritis Res Ther 2011; 13:R29. [PMID: 21338515 PMCID: PMC3241373 DOI: 10.1186/ar3257] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 01/16/2011] [Accepted: 02/21/2011] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Ankylosing spondylitis (AS) is a chronic autoimmune disease, and the precise pathogenesis is largely unknown at present. Bone marrow-derived mesenchymal stem cells (BMSCs) with immunosuppressive and anti-inflammatory potential and Th17/Treg cells with a reciprocal relationship regulated by BMSCs have been reported to be involved in some autoimmune disorders. Here we studied the biological and immunological characteristics of BMSCs, the frequency and phenotype of CCR4+CCR6+ Th/Treg cells and their interaction in vitro in AS. METHODS The biological and immunomodulation characteristics of BMSCs were examined by induced multiple-differentiation and two-way mixed peripheral blood mononuclear cell (PBMC) reactions or after stimulation with phytohemagglutinin, respectively. The interactions of BMSCs and PBMCs were detected with a direct-contact co-culturing system. CCR4+CCR6+ Th/Treg cells and surface markers of BMSCs were assayed using flow cytometry. RESULTS The AS-BMSCs at active stage showed normal proliferation, cell viability, surface markers and multiple differentiation characteristics, but significantly reduced immunomodulation potential (decreased 68 ± 14%); the frequencies of Treg and Fox-P3+ cells in AS-PBMCs decreased, while CCR4+CCR6+ Th cells increased, compared with healthy donors. Moreover, the AS-BMSCs induced imbalance in the ratio of CCR4+CCR6+ Th/Treg cells by reducing Treg/PBMCs and increasing CCR4+CCR6+ Th/PBMCs, and also reduced Fox-P3+ cells when co-cultured with PBMCs. Correlation analysis showed that the immunomodulation potential of BMSCs has significant negative correlations with the ratio of CCR4+CCR6+ Th to Treg cells in peripheral blood. CONCLUSIONS The immunomodulation potential of BMSCs is reduced and the ratio of CCR4+CCR6+ Th/Treg cells is imbalanced in AS. The BMSCs with reduced immunomodulation potential may play a novel role in AS pathogenesis by inducing CCR4+CCR6+ Th/Treg cell imbalance.
Collapse
Affiliation(s)
- Yanfeng Wu
- Department of Othopaedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| | - Mingliang Ren
- Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| | - Rui Yang
- Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| | - Xinjun Liang
- Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| | - Yuanchen Ma
- Department of Othopaedics, Guangdong Provincial People Hospital, 106# Zhongshan Road 2, Guangzhou 510080, PR China
| | - Yong Tang
- Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| | - Lin Huang
- Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| | - Jichao Ye
- Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| | - Keng Chen
- Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| | - Peng Wang
- Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| | - Huiyong Shen
- Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yanjiangxi Road, Guangzhou 510120, PR China
| |
Collapse
|
28
|
Bunnell BA, Betancourt AM, Sullivan DE. New concepts on the immune modulation mediated by mesenchymal stem cells. Stem Cell Res Ther 2010; 1:34. [PMID: 21092149 PMCID: PMC3025436 DOI: 10.1186/scrt34] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are the nonhematopoietic multipotent progenitor cells found in various adult tissues. They are characterized by their ease of isolation and their rapid growth in vitro while maintaining their differentiation potential, allowing for extensive expansion in culture that yields large quantities suitable for therapeutic use. This article reviews the immunomodulatory activities associated with MSCs. Numerous studies have demonstrated that MSCs are potently immunosuppressive in vitro and in vivo. However, this article presents a new paradigm in MSC biology, in which MSCs, at least in vitro, can undergo polarization into either a pro-inflammatory or an immunosuppressive phenotype.
Collapse
Affiliation(s)
- Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, 1430 Tulane Ave, SL-99, New Orleans, LA 70112, USA.
| | | | | |
Collapse
|
29
|
Yu J, Zheng C, Ren X, Li J, Liu M, Zhang L, Liang L, Du W, Han ZC. Intravenous administration of bone marrow mesenchymal stem cells benefits experimental autoimmune myasthenia gravis mice through an immunomodulatory action. Scand J Immunol 2010; 72:242-9. [PMID: 20696022 DOI: 10.1111/j.1365-3083.2010.02445.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSC) are potent in immunomodulation. It has been proven that MSC functioned to correct immune disorder in several immune diseases. Here, we tested the hypothesis that MSC from human bone marrow (hMSC) can provide a potential therapy for experimental autoimmune myasthenia gravis (EAMG). EAMG mice model was established by subcutaneous injection of synthetic analogue of acetylcholine receptor (AchR), then, hMSC were intravenously delivered into these mice repeatedly. The results showed that hMSC could specifically home to spleen tissue and hMSC treatment significantly improved the functional deficits of EAMG mice. In addition, AchR antibody level was dramatically decreased in cell-treated group when compared with untreated control on 10 days after the second cell injection. Moreover, both in vivo and in vitro mixed lymphocyte proliferation assays revealed that hMSC could definitely inhibit the proliferation of AchR-specific lymphocyte. In conclusion, our study demonstrated that hMSC treatment was therapeutically useful in autoimmune myasthenia gravis mice, and the underlying mechanism may relate with their immunomodulatory potential.
Collapse
Affiliation(s)
- J Yu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union, Medical College (CAMS&PUMC), Tianjin, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Söderlund J, Erhardt S, Kast RE. Acyclovir inhibition of IDO to decrease Tregs as a glioblastoma treatment adjunct. J Neuroinflammation 2010; 7:44. [PMID: 20691089 PMCID: PMC2925358 DOI: 10.1186/1742-2094-7-44] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 08/06/2010] [Indexed: 11/16/2022] Open
Abstract
Regulatory T cells, Tregs, are a subset of lymphocytes that have immunosuppressive attributes. They are elevated in blood of glioblastoma patients and within this tumor's tissue itself. Indoleamine 2,3-dioxygenase, IDO, converts tryptophan to kynurenine. IDO activity enhances Treg formation by pathways that are unknown. Experimentally, inhibition of IDO decreases Treg function and number in rodents. The common anti-viral agent acyclovir inhibits IDO. Acyclovir may thereby decrease Treg function in glioblastoma. If it can be confirmed that Treg counts are elevated in glioblastoma patients' tumor tissue, and if we can document acyclovir's lowering of tissue Treg counts by a small trial of acyclovir in pre-operative glioblastoma patients, a trial of acyclovir effect on survival should be done given the current poor prognosis of glioblastoma and the well-established safety and low side effect burden of acyclovir.
Collapse
Affiliation(s)
- Johan Söderlund
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
31
|
Wekerle H, Hohlfeld R. Zero tolerance (to acetylcholine receptor) and ways to overcome it. Ann Neurol 2010; 67:422-4. [PMID: 20437576 DOI: 10.1002/ana.22025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
32
|
Chai LYA, van de Veerdonk F, Marijnissen RJ, Cheng SC, Khoo AL, Hectors M, Lagrou K, Vonk AG, Maertens J, Joosten LAB, Kullberg BJ, Netea MG. Anti-Aspergillus human host defence relies on type 1 T helper (Th1), rather than type 17 T helper (Th17), cellular immunity. Immunology 2010; 130:46-54. [PMID: 20002791 PMCID: PMC2855792 DOI: 10.1111/j.1365-2567.2009.03211.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 10/15/2009] [Accepted: 11/02/2009] [Indexed: 01/22/2023] Open
Abstract
Both interferon-gamma-producing type 1 T helper (Th1)- and interleukin-17 (IL-17)-producing Th17 cells have been proposed to be involved in anti-fungal host defence. Although invasive aspergillosis is one of the most severe human fungal infections, little is known regarding the relative importance of the Th1 versus Th17 cellular immune pathways for the human anti-Aspergillus host defence. Using human peripheral blood mononuclear cells and a system consisting of monocyte-derived macrophages with lymphocytes, we found that Aspergillus fumigatus is a weak inducer of human IL-17 but induces a strong Th1 response. These data were validated by the very low IL-17 levels in bronchoalveolar lavage fluid and serum of patients with invasive aspergillosis. Surprisingly, live A. fumigatus reduced IL-17 production induced by mitogenic stimuli. This effect was mediated through the propensity of A. fumigatus to metabolize tryptophan and release kynurenine, which modulates the inflammatory response through inhibition of IL-17 production. In conclusion, A. fumigatus does not stimulate production of IL-17 and human host defence against aspergillosis may not rely on potent Th17 responses.
Collapse
Affiliation(s)
- Louis Y A Chai
- Department of Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Myasthenia gravis (MG) is an autoimmune neuromuscular junction disease mediated by antibodies against the acetylcholine receptor (AChR). The etiology and immunopathogenesis of MG remain unclear. Recent research has shown the involvement of autoantibodies, lymphocytes, cytokines and chemokines, in the pathogenesis of MG. Systematic factors are also demonstrated, such as inheritance and endocrine. This review indicates the research development in immunopathogenesis of MG.
Collapse
Affiliation(s)
- Sha Huang
- The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | | |
Collapse
|
34
|
Souroujon MC, Brenner T, Fuchs S. Development of novel therapies for MG: Studies in animal models. Autoimmunity 2010; 43:446-60. [DOI: 10.3109/08916930903518081] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|