1
|
Vida H, Sahar M, Nikdouz A, Arezoo H. Chemokines in neurodegenerative diseases. Immunol Cell Biol 2024. [PMID: 39723647 DOI: 10.1111/imcb.12843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/09/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
Neurodegeneration and neuroinflammation disorders are mainly the result of the deposition of various proteins, such as α-synuclein, amyloid-β and prions, which lead to the initiation and activation of inflammatory responses. Different chemokines are involved in the infiltration and movement of inflammatory leukocytes into the central nervous system (CNS) that express chemokine receptors. Dysregulation of several members of chemokines has been shown in the CNS, cerebrospinal fluid and peripheral blood of patients who have neurodegenerative disorders. Upon infiltration of various cells, they produce many inflammatory mediators such as cytokines. Besides them, some CNS-resident cells, such as neurons and astrocytes, are also involved in the pathogenesis of neurodegeneration by producing chemokines. In this review, we summarize the role of chemokines and their related receptors in the pathogenesis of neurodegeneration and neuroinflammation disorders, including multiple sclerosis, Parkinson's disease and Alzheimer's disease. Therapeutic strategies targeting chemokines or their related receptors are also discussed in this article.
Collapse
Affiliation(s)
- Hashemi Vida
- Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mehranfar Sahar
- Cellular and Molecular Medicine Research Institute, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Urmia University of Medical Sciences, Urmia, Iran
| | - Amin Nikdouz
- Department of Translational Medicine, Universita degli Studi del Piemonte Orientale Amedeo Avogadro, Vercelli, Italy
| | - Hosseini Arezoo
- Cellular and Molecular Medicine Research Institute, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
2
|
Toader C, Tataru CP, Munteanu O, Covache-Busuioc RA, Serban M, Ciurea AV, Enyedi M. Revolutionizing Neuroimmunology: Unraveling Immune Dynamics and Therapeutic Innovations in CNS Disorders. Int J Mol Sci 2024; 25:13614. [PMID: 39769374 PMCID: PMC11728275 DOI: 10.3390/ijms252413614] [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: 12/02/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Neuroimmunology is reshaping the understanding of the central nervous system (CNS), revealing it as an active immune organ rather than an isolated structure. This review delves into the unprecedented discoveries transforming the field, including the emerging roles of microglia, astrocytes, and the blood-brain barrier (BBB) in orchestrating neuroimmune dynamics. Highlighting their dual roles in both repair and disease progression, we uncover how these elements contribute to the intricate pathophysiology of neurodegenerative diseases, cerebrovascular conditions, and CNS tumors. Novel insights into microglial priming, astrocytic cytokine networks, and meningeal lymphatics challenge the conventional paradigms of immune privilege, offering fresh perspectives on disease mechanisms. This work introduces groundbreaking therapeutic innovations, from precision immunotherapies to the controlled modulation of the BBB using nanotechnology and focused ultrasound. Moreover, we explore the fusion of immune modulation with neuromodulatory technologies, underscoring new frontiers for personalized medicine in previously intractable diseases. By synthesizing these advancements, we propose a transformative framework that integrates cutting-edge research with clinical translation, charting a bold path toward redefining CNS disease management in the era of precision neuroimmunology.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (M.S.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Calin Petru Tataru
- Department of Opthamology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Central Military Emergency Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Octavian Munteanu
- Department of Anatomy, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (M.S.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Matei Serban
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (M.S.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (M.S.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
- Medical Section, Romanian Academy, 010071 Bucharest, Romania
| | - Mihaly Enyedi
- Department of Anatomy, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
3
|
Taheri M, Tehrani HA, Farzad SA, Korourian A, Arefian E, Ramezani M. The potential of mesenchymal stem cell coexpressing cytosine deaminase and secretory IL18-FC chimeric cytokine in suppressing glioblastoma recurrence. Int Immunopharmacol 2024; 142:113048. [PMID: 39236459 DOI: 10.1016/j.intimp.2024.113048] [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: 05/09/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Glioblastoma multiforme (GBM) patients have a high recurrence rate of 90%, and the 5-year survival rate is only about 5%. Cytosine deaminase (CDA)/5-fluorocytosine (5-FC) gene therapy is a promising glioma treatment as 5-FC can cross the blood-brain barrier (BBB), while 5-fluorouracil (5-FU) cannot. Furthermore, 5-FU can assist reversing the immunological status of cold solid tumors. This study developed mesenchymal stem cells (MSCs) co-expressing yeast CDA and the secretory IL18-FC superkine to prevent recurrent tumor progression by simultaneously exerting cytotoxic effects and enhancing immune responses. IL18 was fused with Igk and IgG2a FC domains to enhance its secretion and serum half-life. The study confirmed the expression and activity of the CDA enzyme, as well as the expression, secretion, and activity of secretory IL18 and IL18-FC superkine, which were expressed by lentiviruses transduced-MSCs. In the transwell tumor-tropism assay, it was observed that the genetically modified MSCs retained their selective tumor-tropism ability following transduction. CDA-expressing MSCs, in the presence of 5-FC (200 µg/ml), induced cell cycle arrest and apoptosis in glioma cells through bystander effects in an indirect transwell co-culture system. They reduced the viability of the direct co-culture system when they constituted only 12.5 % of the cell population. The effectiveness of engineered MSCs in suppressing tumor progression was assessed by intracerebral administration of a lethal dose of GL261 cells combined in a ratio of 1:1 with MSCs expressing CDA, or CDA and sIL18, or CDA and sIL18-FC, into C57BL/6 mice. PET scan showed no conspicuous tumor mass in the MSC-CDA-sIL18-FC group that received 5-FC treatment. The pathological analysis showed that tumor progression suppressed in this group until 20th day after cell inoculation. Cytokine assessment showed that both interferon-gamma (IFN-γ) and interleukin-4 (IL-4) increased in the serum of MSC-CDA-sIL18 and MSC-CDA-sIL18-FC, treated with normal saline (NS) compared to those of the control group. The MSC-CDA-sIL18-FC group that received 5-FC treatment showed reduced serum levels of IL-6 and a considerably improved survival rate compared to the control group. Therefore, MSCs co-expressing yeast CDA and secretory IL18-FC, with tumor tropism capability, may serve as a supplementary approach to standard GBM treatment to effectively inhibit tumor progression and prevent recurrence.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sara Amel Farzad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Korourian
- Quality Control Department Pathobiology Laboratory Center, Tehran, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Stem Cells Technology and Tissue Regeneration Department, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Shimizu F, Nakamori M. Blood-Brain Barrier Disruption in Neuroimmunological Disease. Int J Mol Sci 2024; 25:10625. [PMID: 39408955 PMCID: PMC11476930 DOI: 10.3390/ijms251910625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The blood-brain barrier (BBB) acts as a structural and functional barrier for brain homeostasis. This review highlights the pathological contribution of BBB dysfunction to neuroimmunological diseases, including multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD), autoimmune encephalitis (AE), and paraneoplastic neurological syndrome (PNS). The transmigration of massive lymphocytes across the BBB caused by the activation of cell adhesion molecules is involved in the early phase of MS, and dysfunction of the cortical BBB is associated with the atrophy of gray matter in the late phase of MS. At the onset of NMOSD, increased permeability of the BBB causes the entry of circulating AQP4 autoantibodies into the central nervous system (CNS). Recent reports have shown the importance of glucose-regulated protein (GRP) autoantibodies as BBB-reactive autoantibodies in NMOSD, which induce antibody-mediated BBB dysfunction. BBB breakdown has also been observed in MOGAD, NPSLE, and AE with anti-NMDAR antibodies. Our recent report demonstrated the presence of GRP78 autoantibodies in patients with MOGAD and the molecular mechanism responsible for GRP78 autoantibody-mediated BBB impairment. Disruption of the BBB may explain the symptoms in the brain and cerebellum in the development of PNS, as it induces the entry of pathogenic autoantibodies or lymphocytes into the CNS through autoimmunity against tumors in the periphery. GRP78 autoantibodies were detected in paraneoplastic cerebellar degeneration and Lambert-Eaton myasthenic syndrome, and they were associated with cerebellar ataxia with anti-P/Q type voltage-gated calcium channel antibodies. This review reports that therapies affecting the BBB that are currently available for disease-modifying therapies for neuroimmunological diseases have the potential to prevent BBB damage.
Collapse
Affiliation(s)
- Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan;
| | | |
Collapse
|
5
|
De Masi R, Orlando S, Carata E, Panzarini E. Ultrastructural Characterization of PBMCs and Extracellular Vesicles in Multiple Sclerosis: A Pilot Study. Int J Mol Sci 2024; 25:6867. [PMID: 38999977 PMCID: PMC11241448 DOI: 10.3390/ijms25136867] [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: 05/03/2024] [Revised: 06/10/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Growing evidence identifies extracellular vesicles (EVs) as important cell-to-cell signal transducers in autoimmune disorders, including multiple sclerosis (MS). If the etiology of MS still remains unknown, its molecular physiology has been well studied, indicating peripheral blood mononuclear cells (PBMCs) as the main pathologically relevant contributors to the disease and to neuroinflammation. Recently, several studies have suggested the involvement of EVs as key mediators of neuroimmune crosstalk in central nervous system (CNS) autoimmunity. To assess the role of EVs in MS, we applied electron microscopy (EM) techniques and Western blot analysis to study the morphology and content of plasma-derived EVs as well as the ultrastructure of PBMCs, considering four MS patients and four healthy controls. Through its exploratory nature, our study was able to detect significant differences between groups. Pseudopods and large vesicles were more numerous at the plasmalemma interface of cases, as were endoplasmic vesicles, resulting in an activated aspect of the PBMCs. Moreover, PBMCs from MS patients also showed an increased number of multivesicular bodies within the cytoplasm and amorphous material around the vesicles. In addition, we observed a high number of plasma-membrane-covered extensions, with multiple associated large vesicles and numerous autophagosomal vacuoles containing undigested cytoplasmic material. Finally, the study of EV cargo evidenced a number of dysregulated molecules in MS patients, including GANAB, IFI35, Cortactin, Septin 2, Cofilin 1, and ARHGDIA, that serve as inflammatory signals in a context of altered vesicular dynamics. We concluded that EM coupled with Western blot analysis applied to PBMCs and vesiculation can enhance our knowledge in the physiopathology of MS.
Collapse
Affiliation(s)
- Roberto De Masi
- Complex Operative Unit of Neurology, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| | - Stefania Orlando
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| | - Elisabetta Carata
- Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of the Salento, 73100 Lecce, Italy;
| | - Elisa Panzarini
- Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of the Salento, 73100 Lecce, Italy;
| |
Collapse
|
6
|
Xu M, Wang H, Ren S, Wang B, Yang W, Lv L, Sha X, Li W, Wang Y. Identification of crucial inflammaging related risk factors in multiple sclerosis. Front Mol Neurosci 2024; 17:1398665. [PMID: 38836117 PMCID: PMC11148336 DOI: 10.3389/fnmol.2024.1398665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Background Multiple sclerosis (MS) is an immune-mediated disease characterized by inflammatory demyelinating lesions in the central nervous system. Studies have shown that the inflammation is vital to both the onset and progression of MS, where aging plays a key role in it. However, the potential mechanisms on how aging-related inflammation (inflammaging) promotes MS have not been fully understood. Therefore, there is an urgent need to integrate the underlying mechanisms between inflammaging and MS, where meaningful prediction models are needed. Methods First, both aging and disease models were developed using machine learning methods, respectively. Then, an integrated inflammaging model was used to identify relative risk factors, by identifying essential "aging-inflammation-disease" triples. Finally, a series of bioinformatics analyses (including network analysis, enrichment analysis, sensitivity analysis, and pan-cancer analysis) were further used to explore the potential mechanisms between inflammaging and MS. Results A series of risk factors were identified, such as the protein homeostasis, cellular homeostasis, neurodevelopment and energy metabolism. The inflammaging indices were further validated in different cancer types. Therefore, various risk factors were integrated, and even both the theories of inflammaging and immunosenescence were further confirmed. Conclusion In conclusion, our study systematically investigated the potential relationships between inflammaging and MS through a series of computational approaches, and could present a novel thought for other aging-related diseases.
Collapse
Affiliation(s)
- Mengchu Xu
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Huize Wang
- Department of Nursing, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Siwei Ren
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Bing Wang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Wenyan Yang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Ling Lv
- Department of Thorax, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xianzheng Sha
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Wenya Li
- Department of Thorax, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yin Wang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
7
|
Badaut J, Ghersi-Egea JF, Thorne RG, Konsman JP. Blood-brain borders: a proposal to address limitations of historical blood-brain barrier terminology. Fluids Barriers CNS 2024; 21:3. [PMID: 38183042 PMCID: PMC10770911 DOI: 10.1186/s12987-023-00478-5] [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: 04/11/2023] [Accepted: 10/11/2023] [Indexed: 01/07/2024] Open
Abstract
Many neuroscientists use the term Blood-Brain Barrier (BBB) to emphasize restrictiveness, often equating or reducing the notion of BBB properties to tight junction molecules physically sealing cerebral endothelial cells, rather than pointing out the complexity of this biological interface with respect to its selectivity and variety of exchange between the general blood circulation and the central nervous tissue. Several authors in the field find it unfortunate that the exquisitely dynamic interfaces between blood and brain continue to be viewed primarily as obstructive barriers to transport. Although the term blood-brain interface is an excellent descriptor that does not convey the idea of a barrier, it is important and preferable for the spreading of an idea beyond specialist communities to try to appeal to well-chosen metaphors. Recent evidence reviewed here indicates that blood-brain interfaces are more than selective semi-permeable membranes in that they display many dynamic processes and complex mechanisms for communication. They are thus more like 'geopolitical borders'. Furthermore, some authors working on blood-brain interface-relevant issues have started to use the word border, for example in border-associated macrophages. Therefore, we suggest adopting the term Blood-Brain Border to better communicate the flexibility of and movement across blood-brain interfaces.
Collapse
Affiliation(s)
- Jerome Badaut
- Brain Molecular Imaging Lab, UMR 5536, CNRS, RMSB, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France.
- Basic Science Department, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Jean-François Ghersi-Egea
- FLUID Team, Lyon Neurosciences Research Center, INSERM U1028, CNRS UMR 5292, Lyon-1 University, Bron, France.
| | - Robert G Thorne
- Denali Therapeutics, Inc, 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
- Department of Pharmaceutics, University of Minnesota, 9-177 Weaver-Densford Hall, 308 Harvard St. SE, Minneapolis, MN, 55455, USA.
| | - Jan Pieter Konsman
- UMR 5164, CNRS, ImmunoConcEpT, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France.
| |
Collapse
|
8
|
Yoo KH, Park DJ, Choi JH, Marianayagam NJ, Lim M, Meola A, Chang SD. Optimizing the synergy between stereotactic radiosurgery and immunotherapy for brain metastases. Front Oncol 2023; 13:1223599. [PMID: 37637032 PMCID: PMC10456862 DOI: 10.3389/fonc.2023.1223599] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Solid tumors metastasizing to the brain are a frequent occurrence with an estimated incidence of approximately 30% of all cases. The longstanding conventional standard of care comprises surgical resection and whole-brain radiotherapy (WBRT); however, this approach is associated with limited long-term survival and local control outcomes. Consequently, stereotactic radiosurgery (SRS) has emerged as a potential alternative approach. The primary aim of SRS has been to improve long-term control rates. Nevertheless, rare observations of abscopal or out-of-field effects have sparked interest in the potential to elicit antitumor immunity via the administration of high-dose radiation. The blood-brain barrier (BBB) has traditionally posed a significant challenge to the efficacy of systemic therapy in managing intracranial metastasis. However, recent insights into the immune-brain interface and the development of immunotherapeutic agents have shown promise in preclinical and early-phase clinical trials. Researchers have investigated combining immunotherapy with SRS to enhance treatment outcomes in patients with brain metastasis. The combination approach aims to optimize long-term control and overall survival (OS) outcomes by leveraging the synergistic effects of both therapies. Initial findings have been encouraging in the management of various intracranial metastases, while further studies are required to determine the optimal order of administration, radiation doses, and fractionation regimens that have the potential for the best tumor response. Currently, several clinical trials are underway to assess the safety and efficacy of administering immunotherapeutic agents concurrently or consecutively with SRS. In this review, we conduct a comprehensive analysis of the advantages and drawbacks of integrating immunotherapy into conventional SRS protocols for the treatment of intracranial metastasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Steven D. Chang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
9
|
Górska A, Markiewicz-Gospodarek A, Markiewicz R, Chilimoniuk Z, Borowski B, Trubalski M, Czarnek K. Distribution of Iron, Copper, Zinc and Cadmium in Glia, Their Influence on Glial Cells and Relationship with Neurodegenerative Diseases. Brain Sci 2023; 13:911. [PMID: 37371389 DOI: 10.3390/brainsci13060911] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/30/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Recent data on the distribution and influence of copper, zinc and cadmium in glial cells are summarized. This review also examines the relationship between those metals and their role in neurodegenerative diseases like Alzheimer disease, multiple sclerosis, Parkinson disease and Amyotrophic lateral sclerosis, which have become a great challenge for today's physicians. The studies suggest that among glial cells, iron has the highest concentration in oligodendrocytes, copper in astrocytes and zinc in the glia of hippocampus and cortex. Previous studies have shown neurotoxic effects of copper, iron and manganese, while zinc can have a bidirectional effect, i.e., neurotoxic but also neuroprotective effects depending on the dose and disease state. Recent data point to the association of metals with neurodegeneration through their role in the modulation of protein aggregation. Metals can accumulate in the brain with aging and may be associated with age-related diseases.
Collapse
Affiliation(s)
- Aleksandra Górska
- Department of Human Anatomy, Medical University of Lublin, 4 Jaczewskiego St., 20-090 Lublin, Poland
| | | | - Renata Markiewicz
- Department of Psychiatric Nursing, Medical University of Lublin, 18 Szkolna St., 20-124 Lublin, Poland
| | - Zuzanna Chilimoniuk
- Student Scientific Group at the Department of Family Medicine, 6a (SPSK1) Langiewicza St., 20-032 Lublin, Poland
| | - Bartosz Borowski
- Students Scientific Association at the Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Mateusz Trubalski
- Students Scientific Association at the Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Katarzyna Czarnek
- Institute of Health Sciences, The John Paul II Catholic University of Lublin, Konstantynów 1 H, 20-708 Lublin, Poland
| |
Collapse
|
10
|
Flores-Montoya G, Quintero D, Chatterjea D, Uttley H, Liphart C, Tian Z, Yim E, Hu F. The C-C chemokine receptor 7: An immune molecule that modulates central nervous system function in homeostasis and disease. Brain Behav Immun Health 2023; 29:100610. [PMID: 36937649 PMCID: PMC10015173 DOI: 10.1016/j.bbih.2023.100610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/03/2023] Open
Abstract
The interaction between the central nervous system (CNS) and the peripheral immune system is key for brain function in homeostasis and disease. Recent studies have revealed that the C-C chemokine receptor 7 (CCR7) is expressed in both CNS resident cells and peripheral immune cells, and plays an important role in regulating behavior in homeostasis and neuroinflammation in disease. This review integrates studies examining the role of CCR7 in CNS resident and peripheral immune cells in homeostasis and disease, as well as the pathways of peripheral immune cell migration in and out of the brain via CCR7. A special emphasis is placed on the CCR7-dependent migration of peripheral immune cells into the recently discovered meningeal lymphatic vessels surrounding the brain and nasal lymphatics, its migration into cervical lymph nodes, and the implications that this migration might have for CNS function.
Collapse
Affiliation(s)
- Gisel Flores-Montoya
- Psychology Department, Carleton College, Northfield, MN, USA
- Corresponding author.
| | - Daniel Quintero
- Psychology Department, Carleton College, Northfield, MN, USA
| | | | - Hannah Uttley
- Psychology Department, Carleton College, Northfield, MN, USA
| | - Colin Liphart
- Psychology Department, Carleton College, Northfield, MN, USA
| | - Zichen Tian
- Psychology Department, Carleton College, Northfield, MN, USA
| | - Elliot Yim
- Psychology Department, Carleton College, Northfield, MN, USA
| | - Fengping Hu
- Psychology Department, Carleton College, Northfield, MN, USA
| |
Collapse
|
11
|
Crosstalk between Oxidative Stress and Aging in Neurodegeneration Disorders. Cells 2023; 12:cells12050753. [PMID: 36899889 PMCID: PMC10001353 DOI: 10.3390/cells12050753] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/11/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
The world population is aging rapidly, and increasing lifespan exacerbates the burden of age-related health issues. On the other hand, premature aging has begun to be a problem, with increasing numbers of younger people suffering aging-related symptoms. Advanced aging is caused by a combination of factors: lifestyle, diet, external and internal factors, as well as oxidative stress (OS). Although OS is the most researched aging factor, it is also the least understood. OS is important not only in relation to aging but also due to its strong impact on neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Alzheimer's disease (AD), and Parkinson's disease (PD). In this review, we will discuss the aging process in relation to OS, the function of OS in neurodegenerative disorders, and prospective therapeutics capable of relieving neurodegenerative symptoms associated with the pro-oxidative condition.
Collapse
|
12
|
Hsp65-producing Lactococcus lactis inhibits experimental autoimmune encephalomyelitis by preventing cell migration into spinal cord. Cell Immunol 2023; 384:104661. [PMID: 36621093 DOI: 10.1016/j.cellimm.2022.104661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/19/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023]
Abstract
Multiple sclerosis is an autoimmune disease that affects the central nervous system. Because of its complexity and the difficulty to treat, searching for immunoregulatory responses that reduce the clinical signs of disease by non-aggressive mechanisms and without adverse effects is a scientific challenge. Herein we propose a protocol of oral tolerance induction that prevented and controlled MOG-induced experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice. The genetically modified strain HSP65-producing Lactococcus lactis was orally administered for 5 consecutive days either before or during disease development in mice. Both protocols of feeding HSP65 resulted in significant reduction in the clinical score of EAE. Frequencies of LAP+CD4+Foxp3- regulatory T cells were higher in spleens and inguinal lymph nodes of fed mice. In addition, intravital microscopy showed that adherence of leukocytes to venules in the spinal cord was reduced in orally treated mice. Oral treatment with HSP65-producing L.lactis prevented leukocytes to leave the secondary lymphoid organs, therefore they could not reach the central nervous system. Despite the inhibition of pathological immune response that drive EAE development, activated T cells were at normal frequencies suggesting that oral tolerance did not induce general immunosuppression, but it led to specific control of pathogenic T cells. Our results indicate a novel therapeutic strategy to prevent and control autoimmune diseases such as multiple sclerosis.
Collapse
|
13
|
De Masi R, Orlando S. GANAB and N-Glycans Substrates Are Relevant in Human Physiology, Polycystic Pathology and Multiple Sclerosis: A Review. Int J Mol Sci 2022; 23:7373. [PMID: 35806376 PMCID: PMC9266668 DOI: 10.3390/ijms23137373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Glycans are one of the four fundamental macromolecular components of living matter, and they are highly regulated in the cell. Their functions are metabolic, structural and modulatory. In particular, ER resident N-glycans participate with the Glc3Man9GlcNAc2 highly conserved sequence, in protein folding process, where the physiological balance between glycosylation/deglycosylation on the innermost glucose residue takes place, according GANAB/UGGT concentration ratio. However, under abnormal conditions, the cell adapts to the glucose availability by adopting an aerobic or anaerobic regimen of glycolysis, or to external stimuli through internal or external recognition patterns, so it responds to pathogenic noxa with unfolded protein response (UPR). UPR can affect Multiple Sclerosis (MS) and several neurological and metabolic diseases via the BiP stress sensor, resulting in ATF6, PERK and IRE1 activation. Furthermore, the abnormal GANAB expression has been observed in MS, systemic lupus erythematous, male germinal epithelium and predisposed highly replicating cells of the kidney tubules and bile ducts. The latter is the case of Polycystic Liver Disease (PCLD) and Polycystic Kidney Disease (PCKD), where genetically induced GANAB loss affects polycystin-1 (PC1) and polycystin-2 (PC2), resulting in altered protein quality control and cyst formation phenomenon. Our topics resume the role of glycans in cell physiology, highlighting the N-glycans one, as a substrate of GANAB, which is an emerging key molecule in MS and other human pathologies.
Collapse
Affiliation(s)
- Roberto De Masi
- Complex Operative Unit of Neurology, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy;
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| | - Stefania Orlando
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| |
Collapse
|
14
|
Strickland MR, Alvarez-Breckenridge C, Gainor JF, Brastianos PK. Tumor Immune Microenvironment of Brain Metastases: Toward Unlocking Antitumor Immunity. Cancer Discov 2022; 12:1199-1216. [PMID: 35394521 PMCID: PMC11440428 DOI: 10.1158/2159-8290.cd-21-0976] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/19/2021] [Accepted: 02/17/2022] [Indexed: 11/16/2022]
Abstract
Brain metastasis (BrM) is a devastating complication of solid tumors associated with poor outcomes. Immune-checkpoint inhibitors (ICI) have revolutionized the treatment of cancer, but determinants of response are incompletely understood. Given the rising incidence of BrM, improved understanding of immunobiologic principles unique to the central nervous system (CNS) and dissection of those that govern the activity of ICIs are paramount toward unlocking BrM-specific antitumor immunity. In this review, we seek to discuss the current clinical landscape of ICI activity in the CNS and CNS immunobiology, and we focus, in particular, on the role of glial cells in the CNS immune response to BrM. SIGNIFICANCE There is an urgent need to improve patient selection for and clinical activity of ICIs in patients with cancer with concomitant BrM. Increased understanding of the unique immunobiologic principles that govern response to ICIs in the CNS is critical toward identifying targets in the tumor microenvironment that may potentiate antitumor immunity.
Collapse
Affiliation(s)
| | | | - Justin F Gainor
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
15
|
Benkhoucha M, Tran NL, Breville G, Senoner I, Bradfield PF, Papayannopoulou T, Merkler D, Korn T, Lalive PH. CD4 +c-Met +Itgα4 + T cell subset promotes murine neuroinflammation. J Neuroinflammation 2022; 19:103. [PMID: 35488271 PMCID: PMC9052663 DOI: 10.1186/s12974-022-02461-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/13/2022] [Indexed: 11/21/2022] Open
Abstract
Objective c-Met, a tyrosine kinase receptor, is the unique receptor for hepatocyte growth factor (HGF). The HGF/c-Met axis is reported to modulate cell migration, maturation, cytokine production, and antigen presentation. Here, we report that CD4+c-Met+ T cells are detected at increased levels in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS). Methods c-Met expression by CD4+ T cells was analyzed mostly by flow cytometry and by immunohistochemistry from mice and human PBMCs. The in vivo role of CD4+c-Met+ T cells was assessed in EAE. Results CD4+c-Met+ T cells found in the CNS during EAE peak disease are characterized by a pro-inflammatory phenotype skewed towards a Th1 and Th17 polarization, with enhanced adhesion and transmigration capacities correlating with increased expression of integrin α4 (Itgα4). The adoptive transfer of Itgα4-expressing CD4+Vα3.2+c-Met+ T cells induces increased disease severity compared to CD4+Vα3.2+c-Met− T cells. Finally, CD4+c-Met+ T cells are detected in the brain of MS patients, as well as in the blood with a higher level of Itgα4. These results highlight c-Met as an immune marker of highly pathogenic pro-inflammatory and pro-migratory CD4+ T lymphocytes associated with neuroinflammation. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02461-7.
Collapse
Affiliation(s)
- Mahdia Benkhoucha
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ngoc Lan Tran
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gautier Breville
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Geneva, Switzerland
| | - Isis Senoner
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Paul F Bradfield
- MesenFlow Technologies SARL, Chemin des Aulx 14, Geneva, Switzerland
| | - Thalia Papayannopoulou
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Doron Merkler
- Division of Clinical Pathology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Thomas Korn
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute for Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology, SyNergy, Munich, Germany
| | - Patrice H Lalive
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland. .,Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Geneva, Switzerland.
| |
Collapse
|
16
|
Artemisinin derivative TPN10466 suppresses immune cell migration and Th1/Th17 differentiation to ameliorate disease severity in experimental autoimmune encephalomyelitis. Cell Immunol 2022; 373:104500. [DOI: 10.1016/j.cellimm.2022.104500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/21/2022] [Accepted: 03/02/2022] [Indexed: 11/20/2022]
|
17
|
Matsuyama S, Tanaka Y, Hasebe R, Hojyo S, Murakami M. Gateway Reflex and Mechanotransduction. Front Immunol 2022; 12:780451. [PMID: 35003096 PMCID: PMC8728022 DOI: 10.3389/fimmu.2021.780451] [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] [Received: 09/21/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022] Open
Abstract
The gateway reflex explains how autoreactive CD4+ T cells cause inflammation in tissues that have blood-barriers, such as the central nervous system and retina. It depends on neural activations in response to specific external stimuli, such as gravity, pain, stress, and light, which lead to the secretion of noradrenaline at specific vessels in the tissues. Noradrenaline activates NFkB at these vessels, followed by an increase of chemokine expression as well as a reduction of tight junction molecules to accumulate autoreactive CD4+ T cells, which breach blood-barriers. Transient receptor potential vanilloid 1 (TRPV1) molecules on sensory neurons are critical for the gateway reflex, indicating the importance of mechano-sensing. In this review, we overview the gateway reflex with a special interest in mechanosensory transduction (mechanotransduction).
Collapse
Affiliation(s)
- Shiina Matsuyama
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Tanaka
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Rie Hasebe
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shintaro Hojyo
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Masaaki Murakami
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Chiba, Japan.,Division of Neurommunology, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
18
|
Stamoula Ε, Ainatzoglou A, Stamatellos V, Dardalas I, Siafis S, Matsas A, Stamoulas K, Papazisis G. Atypical antipsychotics in multiple sclerosis: A review of their in vivo immunomodulatory effects. Mult Scler Relat Disord 2022; 58:103522. [DOI: 10.1016/j.msard.2022.103522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/13/2021] [Accepted: 01/09/2022] [Indexed: 11/17/2022]
|
19
|
Lauranzano E, Rasile M, Matteoli M. Integrating Primary Astrocytes in a Microfluidic Model of the Blood-Brain Barrier. Methods Mol Biol 2022; 2492:225-240. [PMID: 35733047 DOI: 10.1007/978-1-0716-2289-6_12] [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] [Indexed: 06/15/2023]
Abstract
An in vitro blood-brain barrier (BBB) model must be highly reproducible and imitate as much as possible the properties of the in vivo environment, from both the functional and anatomical point of view. In our latest work, a BBB prototype was implemented through the use of human primary brain cells and then integrated in a microfluidic platform (Lauranzano et al., Adv Biosyst 3:e1800335, 2019). Here we describe, step by step, the setting of a customized bio-mimetic platform, which uses human brain endothelial cells and primary astrocytic cells to allow the study of the complex interactions between the immune system and the brain in healthy and neuroinflammatory conditions. The model can be exploited to investigate the neuroimmune communication at the blood-brain interface and to examine the transmigration of patient-derived lymphocytes in order to envisage cutting-edge strategies to restore barrier integrity and block the immune cell influx into the CNS.
Collapse
Affiliation(s)
| | - Marco Rasile
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Michela Matteoli
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
- CNR Institute of Neuroscience, Milano, Italy
| |
Collapse
|
20
|
Maternal immune activation with high molecular weight poly(I:C) in Wistar rats leads to elevated immune cell chemoattractants. J Neuroimmunol 2022; 364:577813. [DOI: 10.1016/j.jneuroim.2022.577813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/19/2021] [Accepted: 01/10/2022] [Indexed: 11/20/2022]
|
21
|
Hassanshahi G, Roohi MA, Esmaeili SA, Pourghadamyari H, Nosratabadi R. Involvement of various chemokine/chemokine receptor axes in trafficking and oriented locomotion of mesenchymal stem cells in multiple sclerosis patients. Cytokine 2021; 148:155706. [PMID: 34583254 DOI: 10.1016/j.cyto.2021.155706] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is a specific type of chronic immune-mediated disease in which the immune responses are almost run against the central nervous system (CNS). Despite intensive research, a known treatment for MS disease yet to be introduced. Thus, the development of novel and safe medications needs to be considered for the disease management. Application of mesenchymal stem cells (MSCs) as an emerging approach was recruited forthe treatment of MS. MSCs have several sources and they can be derived from the umbilical cord, adipose tissue, and bone marrow. Chemokines are low molecular weight proteins that their functional activities are achieved by binding to the cell surface G protein-coupled receptors (GPCRs). Chemokine and chemokine receptors are of the most important and effective molecules in MSC trafficking within the different tissues in hemostatic and non-hemostatic circumstances. Chemokine/chemokine receptor axes play a pivotal role in the recruitment and oriented trafficking of immune cells both towards and within the CNS and it appears that chemokine/chemokine receptor signaling may be the most important leading mechanisms in the pathogenesis of MS. In this article, we hypothesized that the chemokine/chemokine receptor axes network have crucial and efficacious impacts on behavior of the MSCs, nonetheless, the exact responsibility of these axes on the targeted tropism of MSCs to the CNS of MS patients yet remained to be fully elucidated. Therefore, we reviewed the ability of MSCs to migrate and home into the CNS of MS patients via expression of various chemokine receptors in response to chemokines expressed by cells of CNS tissue, to provide a great source of knowledge.
Collapse
Affiliation(s)
- Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Amin Roohi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Pourghadamyari
- Department of Clinical Biochemistry, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Reza Nosratabadi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
22
|
A peripheral inflammatory signature discriminates bipolar from unipolar depression: A machine learning approach. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110136. [PMID: 33045321 DOI: 10.1016/j.pnpbp.2020.110136] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/04/2020] [Accepted: 10/06/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Mood disorders (major depressive disorder, MDD, and bipolar disorder, BD) are considered leading causes of life-long disability worldwide, where high rates of no response to treatment or relapse and delays in receiving a proper diagnosis (~60% of depressed BD patients are initially misdiagnosed as MDD) contribute to a growing personal and socio-economic burden. The immune system may represent a new target to develop novel diagnostic and therapeutic procedures but reliable biomarkers still need to be found. METHODS In our study we predicted the differential diagnosis of mood disorders by considering the plasma levels of 54 cytokines, chemokines and growth factors of 81 BD and 127 MDD depressed patients. Clinical diagnoses were predicted also against 32 healthy controls. Elastic net models, including 5000 non-parametric bootstrapping procedure and inner and outer 10-fold nested cross-validation were performed in order to identify the signatures for the disorders. RESULTS Results showed that the immune-inflammatory signature classifies the two disorders with a high accuracy (AUC = 97%), specifically 92% and 86% respectively for MDD and BD. MDD diagnosis was predicted by high levels of markers related to both pro-inflammatory (i.e. IL-1β, IL-6, IL-7, IL-16) and regulatory responses (IL-2, IL-4, and IL-10), whereas BD by high levels of inflammatory markers (CCL3, CCL4, CCL5, CCL11, CCL25, CCL27, CXCL11, IL-9 and TNF-α). CONCLUSIONS Our findings provide novel tools for early diagnosis of BD, strengthening the impact of biomarkers research into clinical practice, and new insights for the development of innovative therapeutic strategies for depressive disorders.
Collapse
|
23
|
Kapoor T, Mehan S. Neuroprotective Methodologies in the Treatment of Multiple Sclerosis Current Status of Clinical and Pre-clinical Findings. Curr Drug Discov Technol 2021; 18:31-46. [PMID: 32031075 DOI: 10.2174/1570163817666200207100903] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/02/2019] [Accepted: 11/26/2019] [Indexed: 11/22/2022]
Abstract
Multiple sclerosis is an idiopathic and autoimmune associated motor neuron disorder that affects myelinated neurons in specific brain regions of young people, especially females. MS is characterized by oligodendrocytes destruction further responsible for demyelination, neuroinflammation, mitochondrial abnormalities, oxidative stress and neurotransmitter deficits associated with motor and cognitive dysfunctions, vertigo and muscle weakness. The limited intervention of pharmacologically active compounds like interferon-β, mitoxantrone, fingolimod and monoclonal antibodies used clinically are majorly associated with adverse drug reactions. Pre-clinically, gliotoxin ethidium bromide mimics the behavioral and neurochemical alterations in multiple sclerosis- like in experimental animals associated with the down-regulation of adenyl cyclase/cAMP/CREB, which is further responsible for a variety of neuropathogenic factors. Despite the considerable investigation of neuroprotection in curing multiple sclerosis, some complications still remain. The available medications only provide symptomatic relief but do not stop the disease progression. In this way, the development of unused beneficial methods tends to be ignored. The limitations of the current steady treatment may be because of their activity at one of the many neurotransmitters included or their failure to up direct signaling flag bearers detailed to have a vital part in neuronal sensitivity, biosynthesis of neurotransmitters and its discharge, development, and separation of the neuron, synaptic versatility and cognitive working. Therefore, the current review strictly focused on the exploration of various clinical and pre-clinical features available for multiple sclerosis to understand the pathogenic mechanisms and to introduce pharmacological interventions associated with the upregulation of intracellular adenyl cyclase/cAMP/CREB activation to ameliorate multiple sclerosis-like features.
Collapse
Affiliation(s)
- Tarun Kapoor
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
24
|
Zhang T, Zhang X, Lin C, Wu S, Wang F, Wang H, Wang Y, Peng Y, Hutchinson MR, Li H, Wang X. Artemisinin inhibits TLR4 signaling by targeting co-receptor MD2 in microglial BV-2 cells and prevents lipopolysaccharide-induced blood-brain barrier leakage in mice. J Neurochem 2021; 157:611-623. [PMID: 33453127 DOI: 10.1111/jnc.15302] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 01/17/2023]
Abstract
Artemisinin and its derivatives have been the frontline drugs for treating malaria. In addition to the antiparasitic effect, accumulating evidence shows that artemisinins can alleviate neuroinflammatory responses in the central nervous system (CNS). However, the precise mechanisms underlying their anti-neuroinflammatory effects are unclear. Herein we attempted to delineate the molecule target of artemisinin in microglia. In vitro protein intrinsic fluorescence titrations and saturation transfer difference (STD)-NMR showed the direct binding of artemisinin to Toll-like receptor TLR4 co-receptor MD2. Cellular thermal shift assay (CETSA) showed that artemisinin binding increased MD2 stability, which implies that artemisinin directly binds to MD2 in the cellular context. Artemisinin bound MD2 showed much less collapse during the molecular dynamic simulations, which supports the increased stability of MD2 upon artemisinin binding. Flow cytometry analysis showed artemisinin inhibited LPS-induced TLR4 dimerization and endocytosis in microglial BV-2 cells. Therefore, artemisinin was found to inhibit the TLR4-JNK signaling axis and block LPS-induced pro-inflammatory factors nitric oxide, IL-1β and TNF-α in BV-2 cells. Furthermore, artemisinin restored LPS-induced decrease of junction proteins ZO-1, Occludin and Claudin-5 in primary brain microvessel endothelial cells, and attenuated LPS-induced blood-brain barrier disruption in mice as assessed by Evans blue. In all, this study unambiguously adds MD2 as a direct binding target of artemisinin in its anti-neuroinflammatory function. The results also suggest that artemisinin could be repurposed as a potential therapeutic intervention for inflammatory CNS diseases.
Collapse
Affiliation(s)
- Tianshu Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China.,Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Yantai University, Yantai, China
| | - Xiaozheng Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Cong Lin
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Siru Wu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Fanfan Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China.,State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences of Guangxi, Normal University, Guilin, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Yibo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Yinghua Peng
- State Key Laboratory for Molecular Biology of Special Economic Animal, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Mark R Hutchinson
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, South Australia, Australia.,ARC Centre of Excellence for Nanoscale Biophotonics, University of Adelaide, Adelaide, SA, Australia
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China.,Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| |
Collapse
|
25
|
Zeitelhofer M, Adzemovic MZ, Moessinger C, Stefanitsch C, Strell C, Muhl L, Brundin L, Fredriksson L, Olsson T, Eriksson U, Nilsson I. Blocking PDGF-CC signaling ameliorates multiple sclerosis-like neuroinflammation by inhibiting disruption of the blood-brain barrier. Sci Rep 2020; 10:22383. [PMID: 33361796 PMCID: PMC7759579 DOI: 10.1038/s41598-020-79598-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Disruption of blood–brain barrier (BBB) integrity is a feature of various neurological disorders. Here we found that the BBB is differently affected during the preclinical, progression and remission phase of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We have identified an upregulation of pro-inflammatory and pro-angiogenic factors in the BBB transcriptome and down-regulation of endothelial tight junction members coinciding with elevated BBB leakage specifically during the progression phase. These changes were antagonized by blocking PDGFRα signaling with the small tyrosine kinase inhibitor imatinib. Moreover, targeting the PDGFRα ligand PDGF-CC using a neutralizing antibody, facilitated recovery of BBB integrity and improvement of EAE symptoms. Intracerebroventricular injection of PDGF-CC induced upregulation, whereas blocking PDGF-CC during EAE led to downregulation of Tnfa and Il1a at the BBB. Our findings suggest that blocking PDGF-CC counteracts fundamental aspects of endothelial cell activation and disruption of the BBB by decreasing Tnfa and Il1a expression. We also demonstrate that both PDGF-CC and its receptor PDGFRα were upregulated in MS lesions indicating that blocking PDGF-CC may be considered a novel treatment for MS.
Collapse
Affiliation(s)
- Manuel Zeitelhofer
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Milena Z Adzemovic
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.,Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Christine Moessinger
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Christina Stefanitsch
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Carina Strell
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden
| | - Lars Muhl
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Lou Brundin
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Linda Fredriksson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Ulf Eriksson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ingrid Nilsson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
26
|
Anchan A, Martin O, Hucklesby JJW, Finlay G, Johnson RH, Robilliard LD, O’Carroll SJ, Angel CE, Graham ES. Analysis of Melanoma Secretome for Factors That Directly Disrupt the Barrier Integrity of Brain Endothelial Cells. Int J Mol Sci 2020; 21:ijms21218193. [PMID: 33139674 PMCID: PMC7663570 DOI: 10.3390/ijms21218193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022] Open
Abstract
We have recently demonstrated that invasive melanoma cells are capable of disrupting the brain endothelial barrier integrity. This was shown using ECIS biosensor technology, which revealed rapid disruption via the paracellular junctions. In this paper, we demonstrate that melanoma cells secrete factors (e.g., cytokines) that weaken the endothelial barrier integrity. Through proteome profiling, we attempt to identify the barrier-disrupting cytokines. Melanoma conditioned media were collected from three New Zealand melanoma lines. ECIS technology was used to assess if the conditioned media disrupted the endothelial barrier independent of the melanoma cells. The melanoma cell secretome was assessed using cytometric bead array (CBA), Luminex immunoassay and multiplex Proteome Profilers, to detect the expression of secretory proteins, which may facilitate metastasis. Finally, ECIS technology was used to assess the direct effects of secreted proteins identified as candidates from the proteome screens. We show that melanoma-conditioned media significantly disrupted the brain endothelial barrier, however, to a much lesser extent than the cells from which they were collected. Cytokine and proteome profiling of the conditioned media showed evidence of high concentrations of approximately 15 secreted proteins (including osteopontin, IL-8, GDF-15, MIF and VEGF). These 15 secreted proteins were expressed variably across the melanoma lines. Surprisingly, the addition of these individually to the brain endothelial cells did not substantially affect the barrier integrity. ANGPTL-4 and TGFβ were also produced by the melanoma cells. Whilst TGFβ-1 had a pronounced effect on the barrier integrity, surprisingly ANGPTL-4 did not. However, its C-terminal fragment did and within a very similar period to the conditioned media, albeit not to the same extent. Herein we show that melanoma cells produce a wide-range of soluble factors at high concentrations, which most likely favour support or survival of the cancer cells. Most of these, except for TGFβ-1 and the C-terminal fragment of ANGPTL-4, did not have an impact on the integrity of the brain endothelial cells.
Collapse
Affiliation(s)
- Akshata Anchan
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (A.A.); (O.M.); (J.J.W.H.); (G.F.); (L.D.R.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (R.H.J.); (S.J.O.)
| | - Olivia Martin
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (A.A.); (O.M.); (J.J.W.H.); (G.F.); (L.D.R.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (R.H.J.); (S.J.O.)
| | - James J. W. Hucklesby
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (A.A.); (O.M.); (J.J.W.H.); (G.F.); (L.D.R.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (R.H.J.); (S.J.O.)
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1010, New Zealand;
| | - Graeme Finlay
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (A.A.); (O.M.); (J.J.W.H.); (G.F.); (L.D.R.)
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Rebecca H. Johnson
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (R.H.J.); (S.J.O.)
- Department of Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Laverne D. Robilliard
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (A.A.); (O.M.); (J.J.W.H.); (G.F.); (L.D.R.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (R.H.J.); (S.J.O.)
| | - Simon J. O’Carroll
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (R.H.J.); (S.J.O.)
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Catherine E. Angel
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1010, New Zealand;
| | - E Scott Graham
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (A.A.); (O.M.); (J.J.W.H.); (G.F.); (L.D.R.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (R.H.J.); (S.J.O.)
- Correspondence:
| |
Collapse
|
27
|
Arneth B. Contributions of T cells in multiple sclerosis: what do we currently know? J Neurol 2020; 268:4587-4593. [PMID: 33083867 DOI: 10.1007/s00415-020-10275-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a complex autoimmune disorder characterized by neurologic dysfunction. The symptoms worsen as the disease progresses to the relapsing stage. AIM This study aimed to examine the role of T cells in MS pathogenesis. MATERIALS AND METHODS The review was performed based on articles obtained from PsycINFO, PubMed, Web of Science, and CINAHL. Search terms and phrases, such as "multiple sclerosis," "MS," "T cells," "development," "Dysregulated T cells," and "Effector T cells", were used to identify articles that could help explore the research topic. RESULTS The pathogenesis of MS is linked to the regulatory, inflammatory, suppressive, and effector roles of T cells. However, the actual roles of specific T cell subsets in MS development are not well understood. DISCUSSION The study revealed a significant link between MS and T cell activity. Targeting T cells is a potential strategy for the development of new therapies to manage MS. CONCLUSION MS is a complex demyelinating condition that affects several million people around the world. Research has revealed that various classes of T cells, including effector T cells and regulatory T cells, influence the development and progression of MS. Further investigations are required to elucidate the underlying mechanisms through which specific T cell populations influence MS pathogenesis.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg UKGM, Justus Liebig University Giessen, Feulgenstr 12, 35440, Giessen, Germany.
| |
Collapse
|
28
|
Sanchez JMS, Doty DJ, DePaula-Silva AB, Brown DG, Bell R, Klag KA, Truong A, Libbey JE, Round JL, Fujinami RS. Molecular patterns from a human gut-derived Lactobacillus strain suppress pathogenic infiltration of leukocytes into the central nervous system. J Neuroinflammation 2020; 17:291. [PMID: 33023618 PMCID: PMC7542353 DOI: 10.1186/s12974-020-01959-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/21/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an inflammatory demyelinating disease that affects 2.5 million people worldwide. Growing evidence suggests that perturbation of the gut microbiota, the dense collection of microorganisms that colonize the gastrointestinal tract, plays a functional role in MS. Indeed, specific gut-resident bacteria are altered in patients with MS compared to healthy individuals, and colonization of gnotobiotic mice with MS-associated microbiota exacerbates preclinical models of MS. However, defining the molecular mechanisms by which gut commensals can remotely affect the neuroinflammatory process remains a critical gap in the field. METHODS We utilized monophasic experimental autoimmune encephalomyelitis (EAE) in C57BL/6J mice and relapse-remitting EAE in SJL/J mice to test the effects of the products from a human gut-derived commensal strain of Lactobacillus paracasei (Lb). RESULTS We report that Lb can ameliorate preclinical murine models of MS with both prophylactic and therapeutic administrations. Lb ameliorates disease through a Toll-like receptor 2-dependent mechanism via its microbe-associated molecular patterns that can be detected in the systemic circulation, are sufficient to downregulate chemokine production, and can reduce immune cell infiltration into the central nervous system (CNS). In addition, alterations in the gut microbiota mediated by Lb-associated molecular patterns are sufficient to provide partial protection against neuroinflammatory diseases. CONCLUSIONS Local Lb modulation of the gut microbiota and the shedding of Lb-associated molecular patterns into the circulation may be important physiological signals to prevent aberrant peripheral immune cell infiltration into the CNS and have relevance to the development of new therapeutic strategies for MS.
Collapse
Affiliation(s)
- John Michael S Sanchez
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Daniel J Doty
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Ana Beatriz DePaula-Silva
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - D Garrett Brown
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Rickesha Bell
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Kendra A Klag
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Amanda Truong
- Department of Oncological Sciences, Huntsman Cancer Institute, 2000 Circle of Hope, 2724 HCI-SOUTH, Salt Lake City, UT, 84112, USA
| | - Jane E Libbey
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - June L Round
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Robert S Fujinami
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
29
|
Hayden L, Semenoff T, Schultz V, Merz SF, Chapple KJ, Rodriguez M, Warrington AE, Shi X, McKimmie CS, Edgar JM, Thümmler K, Linington C, Pingen M. Lipid-specific IgMs induce antiviral responses in the CNS: implications for progressive multifocal leukoencephalopathy in multiple sclerosis. Acta Neuropathol Commun 2020; 8:135. [PMID: 32792006 PMCID: PMC7427287 DOI: 10.1186/s40478-020-01011-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/01/2020] [Indexed: 01/07/2023] Open
Abstract
Progressive multi-focal leukoencephalopathy (PML) is a potentially fatal encephalitis caused by JC polyomavirus (JCV). PML principally affects people with a compromised immune system, such as patients with multiple sclerosis (MS) receiving treatment with natalizumab. However, intrathecal synthesis of lipid-reactive IgM in MS patients is associated with a markedly lower incidence of natalizumab-associated PML compared to those without this antibody repertoire. Here we demonstrate that a subset of lipid-reactive human and murine IgMs induce a functional anti-viral response that inhibits replication of encephalitic Alpha and Orthobunyaviruses in multi-cellular central nervous system cultures. These lipid-specific IgMs trigger microglia to produce IFN-β in a cGAS-STING-dependent manner, which induces an IFN-α/β-receptor 1-dependent antiviral response in glia and neurons. These data identify lipid-reactive IgM as a mediator of anti-viral activity in the nervous system and provide a rational explanation why intrathecal synthesis of lipid-reactive IgM correlates with a reduced incidence of iatrogenic PML in MS.
Collapse
|
30
|
Raviraj R, Nagaraja SS, Selvakumar I, Mohan S, Nagarajan D. The epigenetics of brain tumors and its modulation during radiation: A review. Life Sci 2020; 256:117974. [PMID: 32553924 DOI: 10.1016/j.lfs.2020.117974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/23/2020] [Accepted: 06/10/2020] [Indexed: 10/24/2022]
Abstract
The brain tumor is the abnormal growth of heterogeneous cells around the central nervous system and spinal cord. Most clinically prominent brain tumors affecting both adult and pediatric are glioblastoma, medulloblastoma, and ependymoma and they are classified according to their origin of tissue. Chemotherapy, radiotherapy, and surgery are important treatments available to date. However, these treatments fail due to multiple reasons, including chemoresistance and radiation resistance of cancer cells. Thus, there is a need of new therapeutic designs to target cell signaling and molecular events which are responsible for this resistance. Recently epigenetic changes received increased attention because it helps in understanding chromatin-mediated disease mechanism. The epigenetic modification alters chromatin structure that affects the docking site of many drugs which cause chemo-resistance of cancer therapy. This review centers the mechanism of how epigenetic changes affect the transcription repression and activation of various genes including Polycomb gene, V-Myc avian myelocytomatosis viral oncogene (MYCN). This review also put forth the pathway of radiation-induced reactive oxygen species generation and its role in epigenetic changes in the cellular level and its impact on tissue physiology. Additionally, there is a strong relationship between the behavior of an individual and environment-induced epigenetic regulation of gene expression. The review also discusses Transcriptome heterogeneity and role of tumor microenvironment in glioblastoma. Overall, this review emphasis important and novel epigenetic targets that could be of therapeutic benefit, which helps in overcoming the unsolved chromatin alteration in brain cancer.
Collapse
Affiliation(s)
- Raghavi Raviraj
- Radiation Biology Lab, 206, ASK-II, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu 613401, India
| | - SunilGowda Sunnaghatta Nagaraja
- Radiation Biology Lab, 206, ASK-II, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu 613401, India
| | - Ilakya Selvakumar
- Radiation Biology Lab, 206, ASK-II, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu 613401, India
| | - Suma Mohan
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu 613401, India
| | - Devipriya Nagarajan
- Radiation Biology Lab, 206, ASK-II, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu 613401, India.
| |
Collapse
|
31
|
Yshii L, Bost C, Liblau R. Immunological Bases of Paraneoplastic Cerebellar Degeneration and Therapeutic Implications. Front Immunol 2020; 11:991. [PMID: 32655545 PMCID: PMC7326021 DOI: 10.3389/fimmu.2020.00991] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Paraneoplastic cerebellar degeneration (PCD) is a rare immune-mediated disease that develops mostly in the setting of neoplasia and offers a unique prospect to explore the interplay between tumor immunity and autoimmunity. In PCD, the deleterious adaptive immune response targets self-antigens aberrantly expressed by tumor cells, mostly gynecological cancers, and physiologically expressed by the Purkinje neurons of the cerebellum. Highly specific anti-neuronal antibodies in the serum and cerebrospinal fluid represent key diagnostic biomarkers of PCD. Some anti-neuronal antibodies such as anti-Yo autoantibodies (recognizing the CDR2/CDR2L proteins) are only associated with PCD. Other anti-neuronal antibodies, such as anti-Hu, anti-Ri, and anti-Ma2, are detected in patients with PCD or other types of paraneoplastic neurological manifestations. Importantly, these autoantibodies cannot transfer disease and evidence for a pathogenic role of autoreactive T cells is accumulating. However, the precise mechanisms responsible for disruption of self-tolerance to neuronal self-antigens in the cancer setting and the pathways involved in pathogenesis within the cerebellum remain to be fully deciphered. Although the occurrence of PCD is rare, the risk for such severe complication may increase with wider use of cancer immunotherapy, notably immune checkpoint blockade. Here, we review recent literature pertaining to the pathophysiology of PCD and propose an immune scheme underlying this disabling disease. Additionally, based on observations from patients' samples and on the pre-clinical model we recently developed, we discuss potential therapeutic strategies that could blunt this cerebellum-specific autoimmune disease.
Collapse
Affiliation(s)
- Lidia Yshii
- INSERM U1043, CNRS UMR 5282, Université Toulouse III, Center for Pathophysiology Toulouse Purpan, Toulouse, France
| | - Chloé Bost
- INSERM U1043, CNRS UMR 5282, Université Toulouse III, Center for Pathophysiology Toulouse Purpan, Toulouse, France.,Department of Immunology, Purpan University Hospital Toulouse, Toulouse, France
| | - Roland Liblau
- INSERM U1043, CNRS UMR 5282, Université Toulouse III, Center for Pathophysiology Toulouse Purpan, Toulouse, France.,Department of Immunology, Purpan University Hospital Toulouse, Toulouse, France
| |
Collapse
|
32
|
Yacov N, Kafri P, Salem Y, Propheta-Meiran O, Feldman B, Breitbart E, Mendel I. MOSPD2 is a therapeutic target for the treatment of CNS inflammation. Clin Exp Immunol 2020; 201:105-120. [PMID: 32353176 PMCID: PMC7366745 DOI: 10.1111/cei.13448] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/22/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022] Open
Abstract
In multiple sclerosis and experimental autoimmune encephalomyelitis (EAE), myeloid cells comprise a major part of the inflammatory infiltrate in the central nervous system (CNS). We previously described that motile sperm domain‐containing protein 2 (MOSPD2) is expressed on human myeloid cells and regulates monocyte migration in vitro. The role of MOSPD2 in EAE pathogenesis was studied by generating MOSPD2 knock‐out (KO) mice and monoclonal antibodies directed against MOSPD2. We found that EAE development in MOSPD2 KO mice was significantly suppressed. While frequency representation of leukocyte subsets in lymphoid tissues was comparable, the ratio of inflammatory monocytes in the blood was markedly reduced in MOSPD2 KO mice. In addition, T cells from MOSPD2 KO mice displayed reduced secretion of proinflammatory cytokines and increased production of interleukin (IL)‐4. Prophylactic and post‐onset treatment using monoclonal antibodies (mAbs) generated against MOSPD2 abrogated development and reduced EAE severity. These results suggest that MOSPD2 is key in regulating migration of inflammatory monocytes, and that anti‐MOSPD2 mAbs constitute a potential therapy for the treatment of CNS inflammatory diseases.
Collapse
Affiliation(s)
- N Yacov
- VBL Therapeutics, Modi'in, Israel
| | - P Kafri
- VBL Therapeutics, Modi'in, Israel
| | - Y Salem
- VBL Therapeutics, Modi'in, Israel
| | | | | | | | - I Mendel
- VBL Therapeutics, Modi'in, Israel
| |
Collapse
|
33
|
Sadeghian-Rizi T, Behdani M, Khanahmad H, Sadeghi HM, Jahanian-Najafabadi A. Generation and Characterization of a Functional Nanobody Against Inflammatory Chemokine CXCL10, as a Novel Strategy for the Treatment of Multiple Sclerosis. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2020; 18:141-148. [PMID: 30426906 DOI: 10.2174/1871527317666181114134518] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/21/2018] [Accepted: 10/11/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND & OBJECTIVE Chemokines and their receptors play a pivotal role in the pathogenesis of various autoimmune diseases such as multiple sclerosis, infectious diseases, and also in cancer metastasis via attraction of the pathogenic immune cells into the inflammation sites. METHODS Inflammatory chemokine CXCL10 as a T helper (Th)1-chemokine directs chemotaxis of many cell subsets especially Th1 into the central nervous system (CNS) via its receptor CXCR3 and it has been put forward as a potential therapeutic target in the treatment of multiple sclerosis. Nanobodies are the smallest intact antigen binding fragments derived from heavy chain-only antibodies occurring in camelids with unique biochemical and biophysical features which render them superior to conventional antibodies or antibody fragments. Here, we describe the generation, selection, and characterization of CXCL10-specific Nanobodies from camel immunized with CXCL10. The obtained Nanobodies displayed high affinity towards CXCL10 about 10-11-10-8 M. RESULTS Then a Nanobody with the highest affinity named 3Nb12 was selected and investigated as a migration inhibitor of CXCR3+ cells. Chemotaxis assay results showed that 3Nb12 blocked CXCL10- CXCR3 binding and potently inhibited chemotaxis of CXCR3-transfected HEK293T cells. CONCLUSION The nanobody 3Nb12 might be a promising specific and powerful blocking agent of CXCL10 function, which can be used for diagnostic, therapeutic and research purposes in MS.
Collapse
Affiliation(s)
- Tahereh Sadeghian-Rizi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdi Behdani
- Medical Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Hamid Mirmohammad Sadeghi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
34
|
Robichon K, Patel V, Connor B, La Flamme AC. Clozapine reduces infiltration into the CNS by targeting migration in experimental autoimmune encephalomyelitis. J Neuroinflammation 2020; 17:53. [PMID: 32050980 PMCID: PMC7014621 DOI: 10.1186/s12974-020-01733-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/03/2020] [Indexed: 12/21/2022] Open
Abstract
Background Atypical antipsychotic agents, such as clozapine, are used to treat schizophrenia and other psychiatric disorders by a mechanism that is believed to involve modulating the immune system. Multiple sclerosis is an immune-mediated neurological disease, and recently, clozapine was shown to reduce disease severity in an animal model of MS, experimental autoimmune encephalomyelitis (EAE). However, the mode of action by which clozapine reduces disease in this model is poorly understood. Methods Because the mode of action by which clozapine reduces neuroinflammation is poorly understood, we used the EAE model to elucidate the in vivo and in vitro effects of clozapine. Results In this study, we report that clozapine treatment reduced the infiltration of peripheral immune cells into the central nervous system (CNS) and that this correlated with reduced expression of the chemokines CCL2 and CCL5 transcripts in the brain and spinal cord. We assessed to what extent immune cell populations were affected by clozapine treatment and we found that clozapine targets the expression of chemokines by macrophages and primary microglia. Furthermore, in addition to decreasing CNS infiltration by reducing chemokine expression, we found that clozapine directly inhibits chemokine-induced migration of immune cells. This direct target on the immune cells was not mediated by a change in receptor expression on the immune cell surface but by decreasing downstream signaling via these receptors leading to a reduced migration. Conclusions Taken together, our study indicates that clozapine protects against EAE by two different mechanisms; first, by reducing the chemoattractant proteins in the CNS; and second, by direct targeting the migration potential of peripheral immune cells.
Collapse
Affiliation(s)
- Katharina Robichon
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.,Centre for Biodiscovery Wellington Victoria University of Wellington, Wellington, New Zealand
| | - Vimal Patel
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.,Centre for Biodiscovery Wellington Victoria University of Wellington, Wellington, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Science, University of Auckland, Auckland, New Zealand
| | - Anne Camille La Flamme
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand. .,Centre for Biodiscovery Wellington Victoria University of Wellington, Wellington, New Zealand. .,Malaghan Institute of Medical Research, Wellington, New Zealand.
| |
Collapse
|
35
|
|
36
|
Charabati M, Rabanel JM, Ramassamy C, Prat A. Overcoming the Brain Barriers: From Immune Cells to Nanoparticles. Trends Pharmacol Sci 2019; 41:42-54. [PMID: 31839374 DOI: 10.1016/j.tips.2019.11.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 01/04/2023]
Abstract
Nanoparticulate carriers, often referred to as nanoparticles (NPs), represent an important pharmacological advance for drug protection and tissue-specific drug delivery. Accessing the central nervous system (CNS), however, is a complex process regulated by mainly three brain barriers. While some leukocyte (i.e., immune cell) subsets are equipped with the adequate molecular machinery to infiltrate the CNS in physiological and/or pathological contexts, the successful delivery of NPs into the CNS remains hindered by the tightness of the brain barriers. Here, we present an overview of the three major brain barriers and the mechanisms allowing leukocytes to migrate across each of them. We subsequently review different immune-inspired and -mediated strategies to deliver NPs into the CNS. Finally, we discuss the prospect of exploiting leukocyte trafficking mechanisms for further progress.
Collapse
Affiliation(s)
- Marc Charabati
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Neuroimmunology Unit, Centre de Recherche du CHUM (CRCHUM), Montréal, QC, Canada
| | - Jean-Michel Rabanel
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531, Boulevard des Prairies, Laval, QC, Canada
| | - Charles Ramassamy
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531, Boulevard des Prairies, Laval, QC, Canada.
| | - Alexandre Prat
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Neuroimmunology Unit, Centre de Recherche du CHUM (CRCHUM), Montréal, QC, Canada.
| |
Collapse
|
37
|
QTY code designed thermostable and water-soluble chimeric chemokine receptors with tunable ligand affinity. Proc Natl Acad Sci U S A 2019; 116:25668-25676. [PMID: 31776256 DOI: 10.1073/pnas.1909026116] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chemokine receptors are of great interest as they play a critical role in many immunological and pathological processes. The ability to study chemokine receptors in aqueous solution without detergent would be significant because natural receptors require detergents to become soluble. We previously reported using the QTY code to design detergent-free chemokine receptors. We here report the design of 2 detergent-free chimeric chemokine receptors that were experimentally unattainable in detergent solution. We designed chimeric receptors by switching the N terminus and 3 extracellular (EC) loops between different receptors. Specifically, we replaced the N terminus and 3 EC loops of CCR5QTY with the N terminus and 3 EC loops of CXCR4. The ligand for CXCR4; namely CXCL12, binds to the chimeric receptor CCR5QTY (7TM)-CXCR4 (N terminus+3 EC loops), but with lower affinity compared to CXCR4; the CCL5 ligand of CCR5 binds the chimeric receptor with ∼20× lower affinity. The chimeric design helps to elucidate the mechanism of native receptor-ligand interaction. We also show that all detergent-free QTY-designed chemokine receptors, expressed in Escherichia coli, bind to their respective chemokines with affinities in the nanomolar (nM) range, similar to the affinities of native receptors and SF9-produced QTY variants. These QTY-designed receptors exhibit remarkable thermostability in the presence of arginine and retain ligand-binding activity after heat treatment at 60 °C for 4 h and 24 h, and at 100 °C for 10 min. Our design approach enables affordable scale-up production of detergent-free QTY variant chemokine receptors with tunable functionality for various uses.
Collapse
|
38
|
Ng X, Sadeghian M, Heales S, Hargreaves IP. Assessment of Mitochondrial Dysfunction in Experimental Autoimmune Encephalomyelitis (EAE) Models of Multiple Sclerosis. Int J Mol Sci 2019; 20:ijms20204975. [PMID: 31600882 PMCID: PMC6829485 DOI: 10.3390/ijms20204975] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 01/03/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) that involves the autoreactive T-cell attack on axonal myelin sheath. Lesions or plaques formed as a result of repeated damage and repair mechanisms lead to impaired relay of electrical impulses along the nerve, manifesting as clinical symptoms of MS. Evidence from studies in experimental autoimmune encephalomyelitis (EAE) models of MS strongly suggests that mitochondrial dysfunction presents at the onset of disease and throughout the disease course. The aim of this study was to determine if mitochondrial dysfunction occurs before clinical symptoms arise, and whether this is confined to the CNS. EAE was induced in C57B/L6 mice, and citrate synthase and mitochondrial respiratory chain (MRC) complex I–IV activities were assayed at presymptomatic (3 or 10 days post first immunisation (3 or 10 DPI)) and asymptomatic (17 days post first immunisation (17 DPI) time-points in central nervous system (CNS; spinal cord) and peripheral (liver and jaw muscle) tissues. Samples from animals immunised with myelin oligodendrocyte glycoprotein (MOG) as EAE models were compared with control animals immunised with adjuvant (ADJ) only. Significant changes in MOG compared to control ADJ animals in MRC complex I activity occurred only at presymptomatic stages, with an increase in the spinal cord at 10 DPI (87.9%), an increase at 3 DPI (25.6%) and decrease at 10 DPI (22.3%) in the jaw muscle, and an increase in the liver at 10 DPI (71.5%). MRC complex II/III activity changes occurred at presymptomatic and the asymptomatic stages of the disease, with a decrease occurring in the spinal cord at 3 DPI (87.6%) and an increase at 17 DPI (36.7%), increase in the jaw muscle at 10 DPI (25.4%), and an increase at 3 DPI (75.2%) and decrease at 17 DPI (95.7%) in the liver. Citrate synthase activity was also significantly decreased at 10 DPI (27.3%) in the liver. No significant changes were observed in complex IV across all three tissues assayed. Our findings reveal evidence that mitochondrial dysfunction is present at the asymptomatic stages in the EAE model of MS, and that the changes in MRC enzyme activities are tissue-specific and are not confined to the CNS.
Collapse
Affiliation(s)
- Xiulin Ng
- UCL Institute of Neurology, Queen Square, University College London, London WC1N 3BG, UK.
- University Medical Center, 79106 Freiburg im Breisgau, Germany.
| | - Mona Sadeghian
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London WC1N 3BG, UK.
- Global Clinical Development, Actelion, High Wycombe HP12 4DP, UK.
| | - Simon Heales
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
- Neurometabolic Unit, National Hospital, London WC1N 3BG, UK.
| | - Iain P Hargreaves
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
- Neurometabolic Unit, National Hospital, London WC1N 3BG, UK.
- School of Pharmacy and Biomolecular Sciences, Byrom Street, Liverpool John Moores University, Liverpool L3 3AF, UK.
| |
Collapse
|
39
|
Moura RP, Sarmento B. Therapeutic Approaches toward Multiple Sclerosis: Where Do We Stand and Where Are We Headed? ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Rui Pedro Moura
- CESPU – Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde Rua Central de Gandra 1317 4585‐116 Gandra Portugal
| | - Bruno Sarmento
- CESPU – Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde Rua Central de Gandra 1317 4585‐116 Gandra Portugal
- I3S – Instituto de Investigação e Inovação em SaúdeUniversidade do Porto Rua Alfredo Allen 208 4200‐135 Porto Portugal
- INEB – Instituto de Engenharia BiomédicaUniversidade do Porto Rua Alfredo Allen 208 4200‐135 Porto Portugal
| |
Collapse
|
40
|
Tucker T. Fluid dynamics of cerebrospinal venous flow in multiple sclerosis. Med Hypotheses 2019; 131:109255. [DOI: 10.1016/j.mehy.2019.109255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/27/2019] [Accepted: 06/01/2019] [Indexed: 02/06/2023]
|
41
|
Marro BS, Skinner DD, Cheng Y, Grist JJ, Dickey LL, Eckman E, Stone C, Liu L, Ransohoff RM, Lane TE. Disrupted CXCR2 Signaling in Oligodendroglia Lineage Cells Enhances Myelin Repair in a Viral Model of Multiple Sclerosis. J Virol 2019; 93:e00240-19. [PMID: 31243125 PMCID: PMC6714798 DOI: 10.1128/jvi.00240-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/16/2019] [Indexed: 12/14/2022] Open
Abstract
CXCR2 is a chemokine receptor expressed on oligodendroglia that has been implicated in the pathogenesis of neuroinflammatory demyelinating diseases as well as enhancement of the migration, proliferation, and myelin production by oligodendroglia. Using an inducible proteolipid protein (Plp) promoter-driven Cre-loxP recombination system, we were able to assess how timed ablation of Cxcr2 in oligodendroglia affected disease following intracranial infection with the neurotropic JHM strain of mouse hepatitis virus (JHMV). Generation of Plp-Cre-ER(T)::Cxcr2flox/flox transgenic mice (termed Cxcr2-CKO mice) allows for Cxcr2 to be silenced in oligodendrocytes in adult mice following treatment with tamoxifen. Ablation of oligodendroglia Cxcr2 did not influence clinical severity in response to intracranial infection with JHMV. Infiltration of activated T cells or myeloid cells into the central nervous system (CNS) was not affected, nor was the ability to control viral infection. In addition, the severity of demyelination was similar between tamoxifen-treated mice and vehicle-treated controls. Notably, deletion of Cxcr2 resulted in increased remyelination, as assessed by g-ratio (the ratio of the inner axonal diameter to the total outer fiber diameter) calculation, compared to that in vehicle-treated control mice. Collectively, our findings argue that CXCR2 signaling in oligodendroglia is dispensable with regard to contributing to neuroinflammation, but its deletion enhances remyelination in a preclinical model of the human demyelinating disease multiple sclerosis (MS).IMPORTANCE Signaling through the chemokine receptor CXCR2 in oligodendroglia is important for developmental myelination in rodents, while chemical inhibition or nonspecific genetic deletion of CXCR2 appears to augment myelin repair in animal models of the human demyelinating disease multiple sclerosis (MS). To better understand the biology of CXCR2 signaling on oligodendroglia, we generated transgenic mice in which Cxcr2 is selectively ablated in oligodendroglia upon treatment with tamoxifen. Using a viral model of neuroinflammation and demyelination, we demonstrate that genetic silencing of CXCR2 on oligodendroglia did not affect clinical disease, neuroinflammation, or demyelination, yet there was increased remyelination. These findings support and extend previous findings suggesting that targeting CXCR2 may offer a therapeutic avenue for enhancing remyelination in patients with demyelinating diseases.
Collapse
Affiliation(s)
- Brett S Marro
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California, USA
| | - Dominic D Skinner
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Yuting Cheng
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jonathan J Grist
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Laura L Dickey
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Emily Eckman
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Colleen Stone
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Liping Liu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richard M Ransohoff
- Department of Cell Biology, Harvard University School of Medicine, Boston, Massachusetts, USA
| | - Thomas E Lane
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Immunology, Inflammation & Infectious Disease Initiative, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
42
|
Galli E, Hartmann FJ, Schreiner B, Ingelfinger F, Arvaniti E, Diebold M, Mrdjen D, van der Meer F, Krieg C, Nimer FA, Sanderson N, Stadelmann C, Khademi M, Piehl F, Claassen M, Derfuss T, Olsson T, Becher B. GM-CSF and CXCR4 define a T helper cell signature in multiple sclerosis. Nat Med 2019; 25:1290-1300. [PMID: 31332391 PMCID: PMC6689469 DOI: 10.1038/s41591-019-0521-4] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/11/2019] [Indexed: 12/17/2022]
Abstract
Cytokine dysregulation is a central driver of chronic inflammatory diseases such as multiple sclerosis (MS). Here we sought to determine the characteristic cellular and cytokine polarization profile in patients with relapsing-remitting multiple sclerosis (RRMS) by high-dimensional single-cell mass cytometry (CyTOF). Using a combination of neural network-based representation learning algorithms, we identified an expanded T helper cell subset in MS patients, characterized by the expression of GM-CSF and the C-X-C chemokine receptor type 4. This cellular signature, which includes expression of very late antigen 4 (VLA4) in peripheral blood, was also enriched in the central nervous system of RRMS patients. In independent validation cohorts, we confirmed that this cell population is increased in MS patients compared to other inflammatory and non-inflammatory conditions. Lastly, we also found the population to be reduced under effective disease-modifying therapy, suggesting that the identified T cell profile represents a specific therapeutic target in MS.
Collapse
Affiliation(s)
- Edoardo Galli
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Felix J Hartmann
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.,Department of Dermatology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Bettina Schreiner
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.,Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Eirini Arvaniti
- Institute for Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Martin Diebold
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Dunja Mrdjen
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Franziska van der Meer
- Institut für Neuropathologie, Klinik für Neurologie, Universitätsmedizin Göttingen, Gottingen, Germany
| | - Carsten Krieg
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.,Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Faiez Al Nimer
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Nicholas Sanderson
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Christine Stadelmann
- Institut für Neuropathologie, Klinik für Neurologie, Universitätsmedizin Göttingen, Gottingen, Germany
| | - Mohsen Khademi
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Manfred Claassen
- Institute for Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Tobias Derfuss
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
43
|
Fischer HJ, Finck TLK, Pellkofer HL, Reichardt HM, Lühder F. Glucocorticoid Therapy of Multiple Sclerosis Patients Induces Anti-inflammatory Polarization and Increased Chemotaxis of Monocytes. Front Immunol 2019; 10:1200. [PMID: 31191554 PMCID: PMC6549240 DOI: 10.3389/fimmu.2019.01200] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/13/2019] [Indexed: 01/20/2023] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system (CNS), characterized by the infiltration of mononuclear cells into the CNS and a subsequent inflammation of the brain. Monocytes are implicated in disease pathogenesis not only in their function as potential antigen-presenting cells involved in the local reactivation of encephalitogenic T cells but also by independent effector functions contributing to structural damage and disease progression. However, monocytes also have beneficial effects as they can exert anti-inflammatory activity and promote tissue repair. Glucocorticoids (GCs) are widely used to treat acute relapses in MS patients. They act on a variety of cell types but their exact mechanisms of action including their modulation of monocyte function are not fully understood. Here we investigated effects of the therapeutically relevant GC methylprednisolone (MP) on monocytes from healthy individuals and MS patients in vitro and in vivo. The monocyte composition in the blood was different in MS patients compared to healthy individuals, but it was only marginally affected by MP treatment. In contrast, application of MP caused a marked shift toward an anti-inflammatory monocyte phenotype in vitro and in vivo as revealed by an altered gene expression profile. Chemotaxis of monocytes toward CCL2, CCL5, and CX3CL1 was increased in MS patients compared to healthy individuals and further enhanced by MP pulse therapy. Both of these migration-promoting effects were more pronounced in MS patients with an acute relapse than in those with a progressive disease. Interestingly, the pro-migratory GC effect was independent of chemokine receptor levels as exemplified by results obtained for CCR2. Collectively, our findings suggest that GCs polarize monocytes toward an anti-inflammatory phenotype and enhance their migration into the inflamed CNS, endowing them with the capacity to suppress the pathogenic immune response.
Collapse
Affiliation(s)
- Henrike J Fischer
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Goettingen, Göttingen, Germany.,Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Göttingen, Germany
| | - Tobias L K Finck
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Goettingen, Göttingen, Germany
| | - Hannah L Pellkofer
- Department of Neurology, University Medical Center Goettingen, Göttingen, Germany
| | - Holger M Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Göttingen, Germany
| | - Fred Lühder
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Goettingen, Göttingen, Germany
| |
Collapse
|
44
|
Lauranzano E, Campo E, Rasile M, Molteni R, Pizzocri M, Passoni L, Bello L, Pozzi D, Pardi R, Matteoli M, Ruiz-Moreno A. A Microfluidic Human Model of Blood-Brain Barrier Employing Primary Human Astrocytes. ACTA ACUST UNITED AC 2019; 3:e1800335. [PMID: 32648668 DOI: 10.1002/adbi.201800335] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/20/2019] [Indexed: 12/19/2022]
Abstract
The neurovascular unit (NVU) is the most important biological barrier between vascular districts and central nervous system (CNS) parenchyma, which maintains brain homeostasis, protects the CNS from pathogens penetration, and mediates neuroimmune communication. T lymphocytes migration across the blood-brain barrier is heavily affected in different brain diseases, representing a major target for novel drug development. In vitro models of NVU could represent a primary tool to investigate the molecular events occurring at this interface. To move toward the establishment of personalized therapies, a patient-related NVU-model is set, incorporating human primary astrocytes integrated into a microfluidic platform. The model is morphologically and functionally characterized, proving to be an advantageous tool to investigate human T lymphocytes transmigration and thus the efficacy of potential novel drugs affecting this process.
Collapse
Affiliation(s)
- Eliana Lauranzano
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy
| | - Elena Campo
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy
| | - Marco Rasile
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy.,Department of Biomedical Science, Laboratory of Pharmacology and Brain Pathology, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, MI, Italy
| | - Raffaella Molteni
- Division of Immunology, Transplantation and Infectious Diseases, Leukocyte Biology Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Marco Pizzocri
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy
| | - Lorena Passoni
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy
| | - Lorenzo Bello
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy.,Department of Oncology and Hematology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Davide Pozzi
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy.,Department of Biomedical Science, Laboratory of Pharmacology and Brain Pathology, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, MI, Italy
| | - Ruggero Pardi
- Division of Immunology, Transplantation and Infectious Diseases, Leukocyte Biology Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy.,School of Medicine, Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy
| | - Michela Matteoli
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy.,Department of Biomedical Science, Laboratory of Pharmacology and Brain Pathology, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, MI, Italy
| | - Ana Ruiz-Moreno
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy
| |
Collapse
|
45
|
Ligustilide Ameliorates the Permeability of the Blood–Brain Barrier Model In Vitro During Oxygen–Glucose Deprivation Injury Through HIF/VEGF Pathway. J Cardiovasc Pharmacol 2019; 73:316-325. [DOI: 10.1097/fjc.0000000000000664] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
46
|
Bergmann S, Lawler SE, Qu Y, Fadzen CM, Wolfe JM, Regan MS, Pentelute BL, Agar NYR, Cho CF. Blood-brain-barrier organoids for investigating the permeability of CNS therapeutics. Nat Protoc 2019; 13:2827-2843. [PMID: 30382243 DOI: 10.1038/s41596-018-0066-x] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In vitro models of the blood-brain barrier (BBB) are critical tools for the study of BBB transport and the development of drugs that can reach the CNS. Brain endothelial cells grown in culture are often used to model the BBB; however, it is challenging to maintain reproducible BBB properties and function. 'BBB organoids' are obtained following coculture of endothelial cells, pericytes and astrocytes under low-adhesion conditions. These organoids reproduce many features of the BBB, including the expression of tight junctions, molecular transporters and drug efflux pumps, and hence can be used to model drug transport across the BBB. This protocol provides a comprehensive description of the techniques required to culture and maintain BBB organoids. We also describe two separate detection approaches that can be used to analyze drug penetration into the organoids: confocal fluorescence microscopy and mass spectrometry imaging. Using our protocol, BBB organoids can be established within 2-3 d. An additional day is required to analyze drug permeability. The BBB organoid platform represents an accurate, versatile and cost-effective in vitro tool. It can easily be scaled to a high-throughput format, offering a tool for BBB modeling that could accelerate therapeutic discovery for the treatment of various neuropathologies.
Collapse
Affiliation(s)
- Sonja Bergmann
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sean E Lawler
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuan Qu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Colin M Fadzen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Justin M Wolfe
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael S Regan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bradley L Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Choi-Fong Cho
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
47
|
DeStefano JG, Jamieson JJ, Linville RM, Searson PC. Benchmarking in vitro tissue-engineered blood-brain barrier models. Fluids Barriers CNS 2018; 15:32. [PMID: 30514389 PMCID: PMC6280508 DOI: 10.1186/s12987-018-0117-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/11/2018] [Indexed: 12/13/2022] Open
Abstract
The blood–brain barrier (BBB) plays a key role in regulating transport into and out of the brain. With increasing interest in the role of the BBB in health and disease, there have been significant advances in the development of in vitro models. The value of these models to the research community is critically dependent on recapitulating characteristics of the BBB in humans or animal models. However, benchmarking in vitro models is surprisingly difficult since much of our knowledge of the structure and function of the BBB comes from in vitro studies. Here we describe a set of parameters that we consider a starting point for benchmarking and validation. These parameters are associated with structure (ultrastructure, wall shear stress, geometry), microenvironment (basement membrane and extracellular matrix), barrier function (transendothelial electrical resistance, permeability, efflux transport), cell function (expression of BBB markers, turnover), and co-culture with other cell types (astrocytes and pericytes). In suggesting benchmarks, we rely primarily on imaging or direct measurements in humans and animal models.
Collapse
Affiliation(s)
- Jackson G DeStefano
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - John J Jamieson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA. .,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA. .,120 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA.
| |
Collapse
|
48
|
Hussain RZ, Cravens PC, Doelger R, Dentel B, Herndon E, Loof N, Tsai P, Okuda DT, Racke MK, Stüve O. TLR3 agonism re-establishes CNS immune competence during α4-integrin deficiency. Ann Clin Transl Neurol 2018; 5:1543-1561. [PMID: 30564621 PMCID: PMC6292184 DOI: 10.1002/acn3.664] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 09/13/2018] [Accepted: 09/13/2018] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Natalizumab blocks α4-integrin-mediated leukocyte migration into the central nervous system (CNS). It diminishes disease activity in multiple sclerosis (MS), but carries a high risk of progressive multifocal encephalopathy (PML), an opportunistic infection with JV virus that may be prompted by diminished CNS immune surveillance. The initial host response to viral infections entails the synthesis of type I interferons (IFN) upon engagement of TLR3 receptors. We hypothesized that TLR3 agonism reestablishes CNS immune competence in the setting of α4-integrin deficiency. METHOD We generated the conditional knock out mouse strain Mx1.Cre+ α4-integrinfl/fl, in which the α4-integrin gene is ablated upon treatment with the TLR3 agonist poly I:C. Adoptive transfer of purified lymphocytes from poly I:C-treated Mx1.Cre+ α4-integrinfl/fl donors into naive recipients recapitulates immunosuppression under natalizumab. Active experimental autoimmune encephalomyelitis (EAE) in Mx1.Cre+ α4-integrinfl/fl mice treated with poly I:C represents immune-reconstitution. RESULTS Adoptive transfer of T cells from poly I:C treated Mx1.Cre+ α4-integrinfl/fl mice causes minimal EAE. The in vitro migratory capability of CD45+ splenocytes from these mice is reduced. In contrast, actively-induced EAE after poly I:C treatment results in full disease susceptibility of Mx1.Cre+ α4-integrinfl/fl mice, and the number and composition of CNS leukocytes is similar to controls. Extravasation of Evans Blue indicates a compromised blood-brain barrier. Poly I:C treatment results in a 2-fold increase in IFN β transcription in the spinal cord. INTERPRETATION Our data suggest that TLR3 agonism in the setting of relative α4-integrin deficiency can reestablish CNS immune surveillance in an experimental model. This pathway may present a feasible treatment strategy to treat and prevent PML under natalizumab therapy and should be considered for further experimental evaluation in a controlled setting.
Collapse
Affiliation(s)
- Rehana Z. Hussain
- Department of Neurology and NeurotherapeuticsUniversity of Texas Southwestern Medical CenterDallasTexas
| | - Petra C. Cravens
- Department of Neurology and NeurotherapeuticsUniversity of Texas Southwestern Medical CenterDallasTexas
| | - Richard Doelger
- Department of Neurology and NeurotherapeuticsUniversity of Texas Southwestern Medical CenterDallasTexas
| | - Brianne Dentel
- Department of Neurology and NeurotherapeuticsUniversity of Texas Southwestern Medical CenterDallasTexas
| | - Emily Herndon
- Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTexas
| | - Nicolas Loof
- The Moody Foundation Flow Cytometry FacilityChildren's Research InstituteUniversity of Texas Southwestern Medical CenterDallasTexas
| | - Peter Tsai
- Department of Neurology and NeurotherapeuticsUniversity of Texas Southwestern Medical CenterDallasTexas
| | - Darin T. Okuda
- Department of Neurology and NeurotherapeuticsUniversity of Texas Southwestern Medical CenterDallasTexas
| | | | - Olaf Stüve
- Department of Neurology and NeurotherapeuticsUniversity of Texas Southwestern Medical CenterDallasTexas
- Neurology SectionVA North Texas Health Care System, Medical ServiceDallasTexas
- Department of NeurologyKlinikum rechts der IsarTechnische Universität MünchenMunichGermany
| |
Collapse
|
49
|
El Sharkawi FZ, Ali SA, Hegazy MI, Atya HB. The combined effect of IL-17F and CCL20 gene polymorphism in susceptibility to multiple sclerosis in Egypt. Gene 2018; 685:164-169. [PMID: 30399422 DOI: 10.1016/j.gene.2018.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/01/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Levels of CCL20 and its CCR6 receptor are elevated in many autoimmune diseases which help in the recruitment of T helper (Th17) cells to site of inflammation. OBJECTIVES Determine the value of single nucleotide polymorphism of CCL20 (rs6749704) and IL-17F (rs763780) genes and their concomitant effect on the serum CCL20 level and susceptibility to MS in Egyptian patients. SUBJECTS AND METHODS Blood samples were collected from 83 patients and 95 healthy subjects. Serum levels of CCL20 were measured by ELISA. The DNA was analyzed for rs6749704 and rs763780 using Genotyping Taqman assay. RESULTS The mean serum levels of CCL20 in the MS group were significantly higher than healthy group (P < 0.001). Frequencies of CT genotype of rs6749704 in CCL20 gene and C allele in MS patients were significantly higher compared to controls. Also significant increase of rs763780 in IL-17F gene was detected in MS patients. Concomitant polymorphism in both genes in MS patients showed an increase risk to MS rather than individual locus. CONCLUSION CCL20 may play an important role in the pathogenesis of MS. Both allelic variation of (rs6749704) within CCL20 gene and (rs763780) within IL-17F gene can be considered risk factor for development of MS in Egyptian patients.
Collapse
Affiliation(s)
| | - Sahar A Ali
- Biochemistry Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| | - Mohamed I Hegazy
- Neurology Department, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Hanaa B Atya
- Biochemistry Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| |
Collapse
|
50
|
Kostic M, Zivkovic N, Cvetanovic A, Stojanovic I. Granulocyte-macrophage colony-stimulating factor as a mediator of autoimmunity in multiple sclerosis. J Neuroimmunol 2018; 323:1-9. [DOI: 10.1016/j.jneuroim.2018.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/18/2018] [Accepted: 07/03/2018] [Indexed: 12/20/2022]
|