1
|
Omidi A, Zolfaghari A, Mirab SM, Bafghi MH, Khosravi M, Safdari F, Shirani K. Mapping the complexity of multiple sclerosis: a novel perspective on genetic, environmental, and neurobiological insights. Mol Biol Rep 2025; 52:484. [PMID: 40402311 DOI: 10.1007/s11033-025-10572-6] [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: 02/11/2025] [Accepted: 05/02/2025] [Indexed: 05/23/2025]
Abstract
Multiple sclerosis (MS) is the most common chronic demyelinating disease of the central nervous system (CNS) that mainly affects young adults. MS is a neuroinflammatory disease traditionally classified as an autoimmune disorder; however, its exact cause remains unknown. A wide variety of etiology and risk factors have been proposed to contribute, among which genetics and environment are the leading ones. The heterogeneity of MS can be attributed to a variety of factors, including diverse pathobiological mechanisms. In this narrative review, before discussing the most prevalent etiologies of MS and risk factors, we look at the main neurobiological pathways, blood-brain barrier (BBB) breakdown, and glymphatic system dysfunction. Several intrinsic factors, including genetics and epigenetic implications, hormones, immune system dysregulation, age, and microbiome, have definite roles in developing and worsening MS severity. However, external factors like viruses, bacteria, bioclimate impacts, environmental toxins, lifestyle factors, stress, and psychological factors revealed different or controversial impacts on MS disease. On the other hand, some nascent ones, such as intestinal dysbiosis and COVID-19, need to be further experimentally and clinically investigated. Both may contribute to MS by promoting inflammation and triggering autoimmune responses. Although it assumes that more than one factor contributes to MS development, finding the leading underlying cause and, consequently, the probable involvement mechanisms certainly could help take appropriate, efficient, and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Ameneh Omidi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P. O. Box: 14117-13116, Tehran, Iran.
| | - Amin Zolfaghari
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P. O. Box: 14117-13116, Tehran, Iran
| | - S Mohammadhadi Mirab
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P. O. Box: 14117-13116, Tehran, Iran
| | - Maedeh Hasanzadeh Bafghi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P. O. Box: 14117-13116, Tehran, Iran
| | - Masoumeh Khosravi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P. O. Box: 14117-13116, Tehran, Iran
| | - Fatemeh Safdari
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P. O. Box: 14117-13116, Tehran, Iran
| | - Kobra Shirani
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, P. O. Box: 141556153, Tehran, Iran.
| |
Collapse
|
2
|
Nygrén P, Bouhlal J, Jokinen E, Forstén S, Laajala E, Dias D, Adnan-Awad S, Ianevski A, Klievink J, Lähteenmäki H, Kuusanmäki H, Myllymäki M, Kasanen T, Saeed K, Lee DA, iCAN Study Group, Hjorth-Hansen H, Aittokallio T, Dufva O, Mustjoki S. High-throughput drug screening identifies SMAC mimetics as enhancers of NK-cell cytotoxicity in chronic myeloid leukemia. Blood 2025; 145:1670-1686. [PMID: 39792962 PMCID: PMC12000656 DOI: 10.1182/blood.2024025286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/31/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Natural killer (NK) cells have proven to be safe and effective immunotherapies, associated with favorable treatment responses in chronic myeloid leukemia (CML). Augmenting NK-cell function with oncological drugs could improve NK-cell-based immunotherapies. Here, we used a high-throughput drug screen consisting of >500 small-molecule compounds, to systematically evaluate the effects of oncological drugs on primary NK cells against CML cells. We identified second mitochondrially derived activator of caspases (SMAC) mimetics as potent enhancers of NK-cell cytotoxicity in both cell lines and primary patient samples. In contrast, several drug classes, including glucocorticoids and tyrosine kinase inhibitors such as dasatinib, inhibited NK-cell cytotoxicity. Single-cell RNA sequencing revealed drug-induced transcriptomic changes in both NK and target CML cells. SMAC mimetics upregulated NF-κB target genes in NK cells, potentially contributing to their enhanced cytotoxicity. Inhibitory drugs dexamethasone, dasatinib, and sotrastaurin prevented NK-cell transition to an activated state and suppressed the expression of interferon gamma (IFN-γ) by NK cells, thus preventing IFN-γ-mediated target cell transcriptomic response. In conclusion, we discovered that SMAC mimetics sensitize cancer cells to NK-cell-mediated killing, with potential clinical applications especially in patients with advanced phase CML.
Collapse
MESH Headings
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/drug effects
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- High-Throughput Screening Assays
- Cytotoxicity, Immunologic/drug effects
- Antineoplastic Agents/pharmacology
- Mitochondrial Proteins
- Apoptosis Regulatory Proteins
- Cell Line, Tumor
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Petra Nygrén
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Jonas Bouhlal
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Emmi Jokinen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Sofia Forstén
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Essi Laajala
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Diogo Dias
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Shady Adnan-Awad
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Aleksandr Ianevski
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Jay Klievink
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Hanna Lähteenmäki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Mikko Myllymäki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Tiina Kasanen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Khalid Saeed
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Dean A. Lee
- Division of Hematology, Oncology, and Blood and Marrow Transplant, Nationwide Children's Hospital, Columbus, OH
| | - iCAN Study Group
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Division of Hematology, Oncology, and Blood and Marrow Transplant, Nationwide Children's Hospital, Columbus, OH
- Department of Hematology, St. Olavs Hospital, Trondheim, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | | | - Tero Aittokallio
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Olli Dufva
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
Shrestha N, Dee MJ, Chaturvedi P, Leclerc GM, Mathyer M, Dufour C, Arthur L, Becker-Hapak M, Foster M, McClain E, Pena NV, Kage K, Zhu X, George V, Liu B, Egan J, Echeverri C, Wang M, You L, Kong L, Li L, Berrien-Elliott MM, Cooper ML, Fehniger TA, Rhode PR, Wong HC. A "Prime and Expand" strategy using the multifunctional fusion proteins to generate memory-like NK cells for cell therapy. Cancer Immunol Immunother 2024; 73:179. [PMID: 38960949 PMCID: PMC11222348 DOI: 10.1007/s00262-024-03765-8] [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: 03/07/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024]
Abstract
Adoptive cellular therapy (ACT) using memory-like (ML) natural killer (NK) cells, generated through overnight ex vivo activation with IL-12, IL-15, and IL-18, has shown promise for treating hematologic malignancies. We recently reported that a multifunctional fusion molecule, HCW9201, comprising IL-12, IL-15, and IL-18 domains could replace individual cytokines for priming human ML NK cell programming ("Prime" step). However, this approach does not include ex vivo expansion, thereby limiting the ability to test different doses and schedules. Here, we report the design and generation of a multifunctional fusion molecule, HCW9206, consisting of human IL-7, IL-15, and IL-21 cytokines. We observed > 300-fold expansion for HCW9201-primed human NK cells cultured for 14 days with HCW9206 and HCW9101, an IgG1 antibody, recognizing the scaffold domain of HCW9206 ("Expand" step). This expansion was dependent on both HCW9206 cytokines and interactions of the IgG1 mAb with CD16 receptors on NK cells. The resulting "Prime and Expand" ML NK cells exhibited elevated metabolic capacity, stable epigenetic IFNG promoter demethylation, enhanced antitumor activity in vitro and in vivo, and superior persistence in NSG mice. Thus, the "Prime and Expand" strategy represents a simple feeder cell-free approach to streamline manufacturing of clinical-grade ML NK cells to support multidose and off-the-shelf ACT.
Collapse
Affiliation(s)
- Niraj Shrestha
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Michael J Dee
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | | | - Gilles M Leclerc
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | | | | | | | - Michelle Becker-Hapak
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Mark Foster
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Ethan McClain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | | | - Karen Kage
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Xiaoyun Zhu
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Varghese George
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Bai Liu
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Jack Egan
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | | | - Meng Wang
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Lijing You
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Lin Kong
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Liying Li
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Melissa M Berrien-Elliott
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | | | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Peter R Rhode
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA
| | - Hing C Wong
- HCW Biologics Inc., 2929 N. Commerce Parkway, Miramar, FL, 33025, USA.
| |
Collapse
|
4
|
Fu C, Zhang X, Zhang X, Wang D, Han S, Ma Z. Advances in IL-7 Research on Tumour Therapy. Pharmaceuticals (Basel) 2024; 17:415. [PMID: 38675377 PMCID: PMC11054630 DOI: 10.3390/ph17040415] [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: 02/20/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Interleukin-7 (IL-7) is a versatile cytokine that plays a crucial role in regulating the immune system's homeostasis. It is involved in the development, proliferation, and differentiation of B and T cells, as well as being essential for the differentiation and survival of naïve T cells and the production and maintenance of memory T cells. Given its potent biological functions, IL-7 is considered to have the potential to be widely used in the field of anti-tumour immunotherapy. Notably, IL-7 can improve the tumour microenvironment by promoting the development of Th17 cells, which can in turn promote the recruitment of effector T cells and NK cells. In addition, IL-7 can also down-regulate the expression of tumour growth factor-β and inhibit immunosuppression to promote anti-tumour efficacy, suggesting potential clinical applications for anti-tumour immunotherapy. This review aims to discuss the origin of IL-7 and its receptor IL-7R, its anti-tumour mechanism, and the recent advances in the application of IL-7 in tumour therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhenghai Ma
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (C.F.); (X.Z.); (X.Z.); (D.W.); (S.H.)
| |
Collapse
|
5
|
Chen D, Tang TX, Deng H, Yang XP, Tang ZH. Interleukin-7 Biology and Its Effects on Immune Cells: Mediator of Generation, Differentiation, Survival, and Homeostasis. Front Immunol 2021; 12:747324. [PMID: 34925323 PMCID: PMC8674869 DOI: 10.3389/fimmu.2021.747324] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
Interleukin-7 (IL-7), a molecule known for its growth-promoting effects on progenitors of B cells, remains one of the most extensively studied cytokines. It plays a vital role in health maintenance and disease prevention, and the congenital deficiency of IL-7 signaling leads to profound immunodeficiency. IL-7 contributes to host defense by regulating the development and homeostasis of immune cells, including T lymphocytes, B lymphocytes, and natural killer (NK) cells. Clinical trials of recombinant IL-7 have demonstrated safety and potent immune reconstitution effects. In this article, we discuss IL-7 and its functions in immune cell development, drawing on a substantial body of knowledge regarding the biology of IL-7. We aim to answer some remaining questions about IL-7, providing insights essential for designing new strategies of immune intervention.
Collapse
Affiliation(s)
- Deng Chen
- Division of Trauma and Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting-Xuan Tang
- Class 1901, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Hai Deng
- Division of Trauma and Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang-Ping Yang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Tang
- Division of Trauma and Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Fathi E, Farahzadi R, Valipour B. Alginate/gelatin encapsulation promotes NK cells differentiation potential of bone marrow resident C-kit + hematopoietic stem cells. Int J Biol Macromol 2021; 177:317-327. [PMID: 33621568 DOI: 10.1016/j.ijbiomac.2021.02.131] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/16/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023]
Abstract
The ability of natural killer (NK) cells to destroy cancerous cells with no prior sensitization has made them attractive candidates for cell therapy. The application of hydrogels must be notified as cell delivery vehicles in cell differentiation. The present study was conducted to investigate the effect of alginate-gelatin encapsulation on NK cell differentiation potential of C-kit+ cells. C-kit+ cells were differentiated to NK cells under both encapsulated and un-encapsulated conditions. Next, the cells were subjected to real-time polymerase chain reaction (PCR) and western blotting for the assessment of their telomere length and protein expressions, respectively. Afterward, culture medium was collected to measure cytokines levels. Thereafter, the differentiated NK cells were co-cultured with Molt-4 cells to investigate the potency of cell apoptosis by Annexin V/PI assay. A significant change was observed in the protein expression of Janus kinase/Signal transducers (JAK/STAT) pathway components. Additionally, the encapsulation caused an increase in the apoptosis of Molt-4 cells and telomere length of NK cells differentiated C-kit+ cells. Therefore, it can be concluded that the effects of encapsulation on NK cell's differentiation of C-kit+ cells could be resulted from the secreted cytokines of interleukin (IL)-2, IL-3, IL-7, and IL-12 as well as the increased telomere length.
Collapse
Affiliation(s)
- Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behnaz Valipour
- Department of Histopathology and Anatomy, Faculty of Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
7
|
Carstensen M, Christensen T, Stilund M, Møller HJ, Petersen EL, Petersen T. Activated monocytes and markers of inflammation in newly diagnosed multiple sclerosis. Immunol Cell Biol 2020; 98:549-562. [PMID: 32253768 PMCID: PMC7496724 DOI: 10.1111/imcb.12337] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/08/2020] [Accepted: 03/29/2020] [Indexed: 12/11/2022]
Abstract
In multiple sclerosis (MS), the inflammation and demyelination of the central nervous system (CNS) develop in distinct ways. This makes diagnosing patients difficult, imperative to initiating early and proper treatment. Several common features exist, among them a profound infiltration of monocytes into the CNS mediating demyelination and tissue destruction. In the periphery, monocytes are divided into three subsets depending on expression of CD14 and CD16, representing different stages of activation and differentiation. To investigate their involvement in MS, peripheral blood mononuclear cells (PBMCs) from 61 patients with incipient, untreated MS and 22 symptomatic control (SC) patients as well as 6 patients with radiologically isolated syndrome (RIS) were characterized ex vivo. In addition, paired serum and cerebrospinal fluid (CSF) samples were analyzed with a panel of biomarkers. In PBMC samples, we demonstrate decreased levels of nonclassical monocytes with a concomitant significant decrease of human endogenous retrovirus (HERV) H3 envelope epitopes on this monocyte subset compared with SC and RIS. The observed HERV expression is present on nonclassical monocytes irrespective of MS and thus presumably a result of the inflammatory activation. For the other surface markers analyzed, we found significantly decreased expression between classical and nonclassical monocytes. In matched samples of CSF a highly significant increase in levels of soluble markers of activation and inflammation is shown, and notably this is not the case for the serum samples. Of the soluble markers investigated, interleukin (IL)‐12/IL‐23p40 had the highest discriminatory power in differentiating patients with MS from SC and RIS, almost comparable to the immunoglobulin G index.
Collapse
Affiliation(s)
- Mikkel Carstensen
- Department of Biomedicine, Aarhus University, Skou Building, Høegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark
| | - Tove Christensen
- Department of Biomedicine, Aarhus University, Skou Building, Høegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark
| | - Morten Stilund
- Department of Biomedicine, Aarhus University, Skou Building, Høegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark.,Department of Neurology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, DK-8200, Aarhus N, Denmark
| | - Holger J Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200, Aarhus N, Denmark
| | - Eva L Petersen
- Department of Biomedicine, Aarhus University, Skou Building, Høegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark
| | - Thor Petersen
- Department of Neurology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, DK-8200, Aarhus N, Denmark
| |
Collapse
|
8
|
Mimpen M, Smolders J, Hupperts R, Damoiseaux J. Natural killer cells in multiple sclerosis: A review. Immunol Lett 2020; 222:1-11. [PMID: 32113900 DOI: 10.1016/j.imlet.2020.02.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
As the most common non-traumatic disabling disease among adolescents, multiple sclerosis (MS) is a devastating neurological inflammatory disease of the central nervous system. Research has not yet fully elucidated its pathogenesis, but it has shown MS to be a complex, multifactorial disease with many interplaying factors. One of these factors, natural killer (NK) cells, lymphocytes of the innate immune system, have recently gained attention due to the effects of daclizumab therapy, causing an expansion of the immunoregulatory subset of NK cells. Since then, NK cells and their relation to MS have been the focus of research, with many new findings being published in the last decade. In this review, NK cells are pictured as potent cytotoxic killers, as well as unique immune-regulators. Additionally, an overview of our current knowledge regarding NK cells in MS is given. The role of NK cells in MS is reviewed in the context of well-established environmental factors and current disease modifying therapies to gain further understanding of the pathogenesis and treatment options in MS.
Collapse
Affiliation(s)
- Max Mimpen
- School for Mental Health and Neuroscience, University of Maastricht, Maastricht The Netherlands
| | - Joost Smolders
- Department of Neurology, Erasmus University Medical Center, Rotterdam The Netherlands; Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam The Netherlands
| | - Raymond Hupperts
- School for Mental Health and Neuroscience, University of Maastricht, Maastricht The Netherlands; Department of Neurology, Zuyderland Medical Center, Sittard The Netherlands
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht The Netherlands.
| |
Collapse
|
9
|
Interleukin-7 promotes lung-resident CD14+ monocytes activity in patients with lung squamous carcinoma. Int Immunopharmacol 2019; 67:202-210. [DOI: 10.1016/j.intimp.2018.12.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/04/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022]
|
10
|
Affiliation(s)
- Qiang Liu
- Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Fu-Dong Shi
- Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA/Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China/Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
11
|
Darlington PJ, Stopnicki B, Touil T, Doucet JS, Fawaz L, Roberts ME, Boivin MN, Arbour N, Freedman MS, Atkins HL, Bar-Or A. Natural Killer Cells Regulate Th17 Cells After Autologous Hematopoietic Stem Cell Transplantation for Relapsing Remitting Multiple Sclerosis. Front Immunol 2018; 9:834. [PMID: 29867923 PMCID: PMC5951114 DOI: 10.3389/fimmu.2018.00834] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/05/2018] [Indexed: 12/25/2022] Open
Abstract
In autoimmunity, the balance of different helper T (Th) cell subsets can influence the tissue damage caused by autoreactive T cells. Pro-inflammatory Th1 and Th17 T cells are implicated as mediators of several human autoimmune conditions such as multiple sclerosis (MS). Autologous hematopoietic stem cell transplantation (aHSCT) has been tested in phase 2 clinical trials for MS patients with aggressive disease. Abrogation of new clinical relapses and brain lesions can be seen after ablative aHSCT, accompanied by significant reductions in Th17, but not Th1, cell populations and activity. The cause of this selective decrease in Th17 cell responses following ablative aHSCT is not completely understood. We identified an increase in the kinetics of natural killer (NK) cell reconstitution, relative to CD4+ T cells, in MS patients post-aHSCT, resulting in an increased NK cell:CD4+ T cell ratio that correlated with the degree of decrease in Th17 responses. Ex vivo removal of NK cells from post-aHSCT peripheral blood mononuclear cells resulted in higher Th17 cell responses, indicating that NK cells can regulate Th17 activity. NK cells were also found to be cytotoxic to memory Th17 cells, and this toxicity is mediated through NKG2D-dependent necrosis. Surprisingly, NK cells induced memory T cells to secrete more IL-17A. This was preceded by an early rise in T cell expression of RORC and IL17A mRNA, and could be blocked with neutralizing antibodies against CD58, a costimulatory receptor expressed on NK cells. Thus, NK cells provide initial co-stimulation that supports the induction of a Th17 response, followed by NKG2D-dependent cytotoxicity that limits these cells. Together these data suggest that rapid reconstitution of NK cells following aHSCT contribute to the suppression of the re-emergence of Th17 cells. This highlights the importance of NK cells in shaping the reconstituting immune system following aHSCT in MS patients.
Collapse
Affiliation(s)
- Peter J Darlington
- Departments of Exercise Science and Biology, PERFORM Centre, Concordia University, Montreal, QC, Canada
| | - Brandon Stopnicki
- Departments of Exercise Science and Biology, PERFORM Centre, Concordia University, Montreal, QC, Canada
| | - Tarik Touil
- Neuroimmunology Unit, McGill University and Montreal Neurological Institute, Montreal, QC, Canada
| | - Jean-Sebastien Doucet
- Neuroimmunology Unit, McGill University and Montreal Neurological Institute, Montreal, QC, Canada
| | - Lama Fawaz
- Neuroimmunology Unit, McGill University and Montreal Neurological Institute, Montreal, QC, Canada
| | - Morgan E Roberts
- Neuroimmunology Unit, McGill University and Montreal Neurological Institute, Montreal, QC, Canada
| | - Marie-Noëlle Boivin
- Neuroimmunology Unit, McGill University and Montreal Neurological Institute, Montreal, QC, Canada.,Clinical Biological Imaging and Genetic Repository, McGill University, Montreal, QC, Canada
| | - Nathalie Arbour
- Department of Neurosciences, Université de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Mark S Freedman
- Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Harold L Atkins
- Blood and Marrow Transplant Program, Ottawa General Hospital, Ottawa, ON, Canada
| | - Amit Bar-Or
- Neuroimmunology Unit, McGill University and Montreal Neurological Institute, Montreal, QC, Canada.,Center for Neuroinflammation and Experimental Therapeutics and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
12
|
Toghraie FS, Sharifzadeh SM, Ramezani A, Mahmoudi Maymand1 E, Yazdanpanah-Samani M, Ghaderi A. Cloning and Expression of Recombinant Human Interleukin-7 in Chinese Hamster Ovary (CHO) Cells. Rep Biochem Mol Biol 2017; 6:66-73. [PMID: 29090231 PMCID: PMC5643452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 02/23/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND The critical role of interleukin-7 (IL-7) in homeostatic proliferation and T cell survival has made it a promising cytokine for the treatment of various clinical conditions, especially those associated with lymphopenia. METHODS In the present study we expressed recombinant human interleukin-7 (rhIL-7) in Chinese hamster ovary (CHO)-K1 cells. CHO-K1 cells were stably transfected with both circular and linear forms of the pBud-hIL-7 recombinant by electroporation. Expression of rhIL-7 in CHO-K1 cells was confirmed by enzyme-linked immunosorbent assay (ELISA) and dot and western blots. RESULTS On western blots of transformed cells, a single 25 kDa band was observed, consistent with the expected molecular weight of glycosylated hIL-7. No significant expression difference was observed between cells transfected with circular or linear plasmids. CONCLUSION We established a stable CHO-K1 cell line expressing rhIL-7, which we consider to be a promising system for the production of rhIL-7 as a biopharmaceutical.
Collapse
Affiliation(s)
- Fatemeh Sadat Toghraie
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical science, Shiraz, Iran.
| | - Seyedeh Masoumeh Sharifzadeh
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical science, Shiraz, Iran.
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical science, Shiraz, Iran.
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Elham Mahmoudi Maymand1
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical science, Shiraz, Iran.
| | - Mahsa Yazdanpanah-Samani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical science, Shiraz, Iran.
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical science, Shiraz, Iran.
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
13
|
Judge CJ, Kostadinova L, Sherman KE, Butt AA, Falck-Ytter Y, Funderburg NT, Landay AL, Lederman MM, Sieg SF, Sandberg JK, Anthony DD. CD56 bright NK IL-7Rα expression negatively associates with HCV level, and IL-7-induced NK function is impaired during HCV and HIV infections. J Leukoc Biol 2017; 102:171-184. [PMID: 28400540 DOI: 10.1189/jlb.5a1116-456r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/12/2017] [Accepted: 03/15/2017] [Indexed: 12/13/2022] Open
Abstract
Several lines of evidence support the concept that NK cells play an important role in control of hepatitis C virus (HCV) infection via cytokine secretion and cytotoxicity. IL-7 is a homeostatic cytokine with a role in T cell development, activation, proliferation, and cytokine secretion. The IL-7Rα chain [cluster of differentiation (CD)127] is expressed on NK cells, with greatest abundance on the CD56brightCD16dim/- (CD56bright) subset. Here, we measured CD127 expression on CD56bright, CD56dimCD16+ (CD56dim), or CD56negCD16+ (CD56neg) NK cell subsets of 25 uninfected donors (UD); 34 chronic HCV-infected, treatment-naïve; 25 HIV-infected, virally suppressed on antiretroviral therapy (ART); and 42 HCV-HIV-coinfected subjects on ART. Interestingly, CD127 expression on CD56bright NK cells negatively correlated with HCV plasma levels in HCV monoinfection and HCV-HIV coinfection. IL-7 induced CD69 expression, as well as IFN-γ production, in CD56bright NK cells and also enhanced the IFN-α-induced CD69 expression on these cells. The latter was impaired in HIV infection. Furthermore, IL-7 induced B cell lymphoma 2 (BCL-2) expression and cell cycling of CD56bright NK cells, and this effect was impaired in HCV- and HIV-infected subjects. Whereas IL-7-stimulated CD56bright NK cell degranulation appeared intact in all cohorts, we observed impaired IL-7-activated NK cell cytolytic function in HCV- and HIV-infected subjects. Finally, IL-7-induced phosphorylation of STAT-5 (pSTAT-5) signaling was impaired in NK cells of subjects with chronic viral infection, and this was reversible upon 6 mo of viral suppression with IFN-free HCV therapy. These results implicate that IL-7-dependent NK cell activation and effector function may be other host immune surveillance mechanisms that are impaired in viral infections.
Collapse
Affiliation(s)
- Chelsey J Judge
- Department of Pathology, Cleveland VA Medical Center, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Medicine, University Hospitals Case Medical Center and Center for AIDS Research (CFAR), Case Western Reserve University, Cleveland, Ohio, USA
| | - Lenche Kostadinova
- Department of Medicine, University Hospitals Case Medical Center and Center for AIDS Research (CFAR), Case Western Reserve University, Cleveland, Ohio, USA
| | - Kenneth E Sherman
- Department of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Adeel A Butt
- Weill Cornell Medical College, New York, New York, USA.,Hamad Healthcare Quality Institute and Hamad Medical Corporation, Doha, Qatar
| | - Yngve Falck-Ytter
- Department of Pathology, Cleveland VA Medical Center, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Medicine, University Hospitals Case Medical Center and Center for AIDS Research (CFAR), Case Western Reserve University, Cleveland, Ohio, USA
| | - Nicholas T Funderburg
- School of Health and Rehabilitation, Division of Medical Laboratory Science, The Ohio State University, Columbus, Ohio, USA
| | - Alan L Landay
- Rush University Medical Center, Chicago, Illinois, USA: and
| | - Michael M Lederman
- Department of Medicine, University Hospitals Case Medical Center and Center for AIDS Research (CFAR), Case Western Reserve University, Cleveland, Ohio, USA
| | - Scott F Sieg
- Department of Medicine, University Hospitals Case Medical Center and Center for AIDS Research (CFAR), Case Western Reserve University, Cleveland, Ohio, USA
| | - Johan K Sandberg
- Center for Infection Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Donald D Anthony
- Department of Pathology, Cleveland VA Medical Center, Case Western Reserve University, Cleveland, Ohio, USA; .,Department of Medicine, University Hospitals Case Medical Center and Center for AIDS Research (CFAR), Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
14
|
Gao M, Yang Y, Li D, Ming B, Chen H, Sun Y, Xiao Y, Lai L, Zou H, Xu Y, Xiong P, Tan Z, Gong F, Zheng F. CD27 natural killer cell subsets play different roles during the pre-onset stage of experimental autoimmune encephalomyelitis. Innate Immun 2016; 22:395-404. [DOI: 10.1177/1753425916658111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/14/2016] [Indexed: 12/28/2022] Open
Abstract
NK cells participate in the development of human multiple sclerosis (MS) and mouse experimental autoimmune encephalomyelitis (EAE), but the roles of different NK cell subsets in disease onset remain poorly understood. In this study, murine NK cells were divided into CD27high and CD27low/− subsets. The CD27high subset was decreased and the CD27low/− subset was increased in lymphoid organs during the pre-onset stage of EAE. Compared with the counterpart in naïve mice, the CD27high subset showed lower expression of Ly49D, Ly49H and NKG2D, and less production of IFN-γ, whereas the CD27low/− subset showed similar expression of the above mentioned surface receptors but higher cytotoxic activity in EAE mice. Compared with the CD27high subset, the CD27low/− subset exhibited increased promotion of DC maturation and no significant inhibition of T cells proliferation and Th17 cells differentiation in vitro. Additionally, adoptive transfer of the CD27low/− subset, but not the CD27high subset, exacerbated the severity of EAE. Collectively, our data suggest the CD27 NK cell subsets play different roles in controlling EAE onset, which provide a new understanding for the regulation of NK cell subsets in early autoimmune disease.
Collapse
Affiliation(s)
- Ming Gao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Yang
- Division of Viral Pathology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Daling Li
- Department of Anesthesiology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingxia Ming
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huoying Chen
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Sun
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Xiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lai
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huijuan Zou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Xu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Xiong
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Tan
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feili Gong
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|