1
|
Kim MW, Gao W, Lichti CF, Gu X, Dykstra T, Cao J, Smirnov I, Boskovic P, Kleverov D, Salvador AFM, Drieu A, Kim K, Blackburn S, Crewe C, Artyomov MN, Unanue ER, Kipnis J. Endogenous self-peptides guard immune privilege of the central nervous system. Nature 2025; 637:176-183. [PMID: 39476864 DOI: 10.1038/s41586-024-08279-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/23/2024] [Indexed: 12/06/2024]
Abstract
Despite the presence of strategically positioned anatomical barriers designed to protect the central nervous system (CNS), it is not entirely isolated from the immune system1,2. In fact, it remains physically connected to, and can be influenced by, the peripheral immune system1. How the CNS retains such responsiveness while maintaining an immunologically unique status remains an outstanding question. Here, in searching for molecular cues that derive from the CNS and enable its direct communication with the immune system, we identified an endogenous repertoire of CNS-derived regulatory self-peptides presented on major histocompatibility complex class II (MHC-II) molecules in the CNS and at its borders. During homeostasis, these regulatory self-peptides were found to be bound to MHC-II molecules throughout the path of lymphatic drainage from the brain to its surrounding meninges and its draining cervical lymph nodes. However, in neuroinflammatory disease, the presentation of regulatory self-peptides diminished. After boosting the presentation of these regulatory self-peptides, a population of suppressor CD4+ T cells was expanded, controlling CNS autoimmunity in a CTLA-4- and TGFβ-dependent manner. CNS-derived regulatory self-peptides may be the molecular key to ensuring a continuous dialogue between the CNS and the immune system while balancing overt autoreactivity. This sheds light on how we conceptually think about and therapeutically target neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Min Woo Kim
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Immunology Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Medical Scientist Training Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Wenqing Gao
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Cheryl F Lichti
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Bursky Center for Human Immunology and Immunotherapy Programs, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Xingxing Gu
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Taitea Dykstra
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Jay Cao
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Igor Smirnov
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Pavle Boskovic
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Denis Kleverov
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Andrea F M Salvador
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Antoine Drieu
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Kyungdeok Kim
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Susan Blackburn
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Clair Crewe
- Department of Cell Biology and Physiology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Bursky Center for Human Immunology and Immunotherapy Programs, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Emil R Unanue
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Bursky Center for Human Immunology and Immunotherapy Programs, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
- Bursky Center for Human Immunology and Immunotherapy Programs, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
2
|
Liu D, Zhu C, Wei H, Xu Q. A mouse model of schizophrenia induced by autoantibodies against SFT2D2. Neuroscience 2024; 558:30-36. [PMID: 39067681 DOI: 10.1016/j.neuroscience.2024.07.033] [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: 05/30/2024] [Revised: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Schizophrenia (SCZ) is a highly heterogeneous, severe neuropsychiatric disorder of unknown etiopathology. Increasing data indicate an overlap between schizophrenia and pathological processes related to immunological dysregulation as well as inflammation, such as high levels of pro-inflammatory substances in patients' blood and cerebrospinal fluid and autoantibodies against synaptic and nerve cell membrane proteins. Autoantibodies against SFT2D2 have been reported in patients with SCZ. However, their roles in inflammation have not yet been established. We performed a continuous intracerebroventricular infusion of polyclonal rabbit anti-SFT2D2-IgG in male C57BL/6 mice. Behavioral tests were conducted after 2 weeks of treatment. Our results showed an increased density of microglia and activated astrocytes in the primary somatosensory cortex of the anti-SFT2D2-IgG-infused mice. Quantitative reverse transcription-polymerase chain reactions showed that the expression of pro-inflammatory genes was upregulated in the primary somatosensory cortex and hippocampus of the anti-SFT2D2-IgG-infused mice. Additionally, the mice exhibited defective sensorimotor gating, memory deficits, motor impairment, and anxiety-related behaviors without signs of depression. These findings indicate that anti-SFT2D2 autoantibodies can induce encephalitis, cause a series of behavioral changes associated with schizophrenia, and offer a model for testing novel therapies to improve treatment strategies for a subgroup of patients with SCZ.
Collapse
Affiliation(s)
- Duilin Liu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Caiyun Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui Wei
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China.
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Kapel-Reguła A, Chojdak-Łukasiewicz J, Rybińska A, Duś-Ilnicka I, Radwan-Oczko M. Oral State and Salivary Cortisol in Multiple Sclerosis Patients. Biomedicines 2024; 12:2277. [PMID: 39457590 PMCID: PMC11504177 DOI: 10.3390/biomedicines12102277] [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: 08/05/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background: MS patients experience gradual and progressive functional limitation, bulbar symptoms, cognitive dysfunction, and psychiatric disorders that can impinge on oral status. This study aimed to investigate the oral state, oral hygiene habits, and salivary cortisol levels in patients with relapsing-remitting multiple sclerosis (RRMS) compared to healthy controls. It also evaluated systemic parameters: disease duration, type of Disease Modifying Therapy (DMT), disability score, professional activity, and smoking in the study group. Methods: This study included 101 patients (71 women and 30 men, aged 16-71 years) and 51 healthy volunteers (36 women and 15 men, aged 28-82 years). The oral examination assessed the number of teeth, type and number of dental fillings and prosthetic restoration, oral hygiene state, and salivary cortisol. Results: It was found that MS patients had significantly more professional activity, swallowing problems, pronunciation issues, dry mouth, and taste disturbances than the control group. They brushed their teeth twice daily significantly less often. The API was higher, while the SBI was lower in MS patients. Disease duration positively correlated with age and number of missing teeth. The Expanded Disability Status Scale positively correlated with age, disease duration, number of missing teeth, number of composite fillings, and right and left-hand Nine Hole Peg test scores, and negatively correlated with the Sulcus Bleeding Index. Salivary cortisol levels did not differ between groups and correlated only with the disability scale. Conclusions: MS patients require ongoing dental care and preventive measures to manage both general and oral health symptoms effectively.
Collapse
Affiliation(s)
| | | | - Anna Rybińska
- Department of Oral Pathology, Faculty of Dentistry, Wroclaw Medical University, 50-425 Wroclaw, Poland; (A.R.); (I.D.-I.); (M.R.-O.)
| | - Irena Duś-Ilnicka
- Department of Oral Pathology, Faculty of Dentistry, Wroclaw Medical University, 50-425 Wroclaw, Poland; (A.R.); (I.D.-I.); (M.R.-O.)
| | - Małgorzata Radwan-Oczko
- Department of Oral Pathology, Faculty of Dentistry, Wroclaw Medical University, 50-425 Wroclaw, Poland; (A.R.); (I.D.-I.); (M.R.-O.)
| |
Collapse
|
4
|
Seida I, Al Shawaf M, Mahroum N. Fecal microbiota transplantation in autoimmune diseases - An extensive paper on a pathogenetic therapy. Autoimmun Rev 2024; 23:103541. [PMID: 38593970 DOI: 10.1016/j.autrev.2024.103541] [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: 01/11/2024] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
The role of infections in the pathogenesis of autoimmune diseases has long been recognized and reported. In addition to infectious agents, the internal composition of the "friendly" living bacteria, (microbiome) and its correlation to immune balance and dysregulation have drawn the attention of researchers for decades. Nevertheless, only recently, scientific papers regarding the potential role of transferring microbiome from healthy donor subjects to patients with autoimmune diseases has been proposed. Fecal microbiota transplantation or FMT, carries the logic of transferring microorganisms responsible for immune balance from healthy donors to individuals with immune dysregulation or more accurately for our paper, autoimmune diseases. Viewing the microbiome as a pathogenetic player allows us to consider FMT as a pathogenetic-based treatment. Promising results alongside improved outcomes have been demonstrated in patients with different autoimmune diseases following FMT. Therefore, in our current extensive review, we aimed to highlight the implication of FMT in various autoimmune diseases, such as inflammatory bowel disease, autoimmune thyroid and liver diseases, systemic lupus erythematosus, and type 1 diabetes mellitus, among others. Presenting all the aspects of FMT in more than 12 autoimmune diseases in one paper, to the best of our knowledge, is the first time presented in medical literature. Viewing FMT as such could contribute to better understanding and newer application of the model in the therapy of autoimmune diseases, indeed.
Collapse
Affiliation(s)
- Isa Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Maisam Al Shawaf
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
5
|
Tatomir A, Anselmo F, Boodhoo D, Chen H, Mekala AP, Nguyen V, Cuevas J, Rus V, Rus H. Multiple sclerosis disease activity, a multi-biomarker score of disease activity and response to treatment in multiple sclerosis. Front Immunol 2024; 15:1338585. [PMID: 38994359 PMCID: PMC11236682 DOI: 10.3389/fimmu.2024.1338585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Regular assessment of disease activity in relapsing-remitting multiple sclerosis (RRMS) is required to optimize clinical outcomes. Biomarkers can be a valuable tool for measuring disease activity in multiple sclerosis (MS) if they reflect the pathological processes underlying MS pathogenicity. In this pilot study, we combined multiple biomarkers previously analyzed in RRMS patients into an MS disease activity (MSDA) score to evaluate their ability to predict relapses and treatment response to glatiramer acetate (GA). Response Gene to Complement 32 (RGC-32), FasL, IL-21, SIRT1, phosphorylated SIRT1 (p-SIRT1), and JNK1 p54 levels were used to generate cut-off values for each biomarker. Any value below the cutoff for RGC-32, FasL SIRT1, or p-SIRT1 or above the cutoff for IL-21 or JNK1 p54 was given a +1 value, indicating relapse or lack of response to GA. Any value above the cutoff value for RGC-32, FasL, SIRT1, p-SIRT1 or below that for IL-21 or JNK1 p54 was given a -1 value, indicating clinical stability or response to GA. An MSDA score above +1 indicated a relapse or lack of response to treatment. An MSDA score below -1 indicated clinical stability or response to treatment. Our results showed that the MSDA scores generated using either four or six biomarkers had a higher sensitivity and specificity and significantly correlated with the expanded disability status scale. Although these results suggest that the MSDA test can be useful for monitoring therapeutic response to biologic agents and assessing clinically challenging situations, the present findings need to be confirmed in larger studies.
Collapse
Affiliation(s)
- Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Neurology Department, Baltimore Veterans Administration Hospital, Baltimore, MD, United States
| | - Freidrich Anselmo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Hegang Chen
- Department of Epidemiology and Public Health, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Armugam P. Mekala
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Jacob Cuevas
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Neurology Department, Baltimore Veterans Administration Hospital, Baltimore, MD, United States
| |
Collapse
|
6
|
Seo D, So JM, Kim J, Jung H, Jang I, Kim H, Kang DW, Lim YM, Choi J, Lee EJ. Digital symbol-digit modalities test with modified flexible protocols in patients with CNS demyelinating diseases. Sci Rep 2024; 14:14649. [PMID: 38918552 PMCID: PMC11199480 DOI: 10.1038/s41598-024-65486-3] [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: 02/02/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024] Open
Abstract
Cognitive impairment (CI) is prevalent in central nervous system demyelinating diseases, such as multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD). We developed a novel tablet-based modified digital Symbol Digit Modalities Test (MD-SDMT) with adjustable protocols that feature alternating symbol-digit combinations in each trial, lasting one or two minutes. We assessed 144 patients (99 with MS and 45 with NMOSD) using both MD-SDMT protocols and the traditional paper-based SDMT. We also gathered participants' feedback through a questionnaire regarding their preferences and perceived reliability. The results showed strong correlations between MD-SDMT and paper-based SDMT scores (Pearsons correlation: 0.88 for 2 min; 0.85 for 1 min, both p < 0.001). Among the 120 respondents, the majority preferred the digitalized SDMT (55% for the 2 min, 39% for the 1 min) over the paper-based version (6%), with the 2 min MD-SDMT reported as the most reliable test. Notably, patients with NMOSD and older individuals exhibited a preference for the paper-based test, as compared to those with MS and younger patients. In summary, even with short test durations, the digitalized SDMT effectively evaluates cognitive function in MS and NMOSD patients, and is generally preferred over the paper-based method, although preferences may vary with patient characteristics.
Collapse
Affiliation(s)
- Dayoung Seo
- AMIST, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Jeong Min So
- Department of Neurology, Asan Medical Center, University of Ulsan, Seoul, 05505, South Korea
| | - Jiyon Kim
- Department of Neurology, Asan Medical Center, University of Ulsan, Seoul, 05505, South Korea
| | - Heejae Jung
- Department of Neurology, Asan Medical Center, University of Ulsan, Seoul, 05505, South Korea
| | - Inhye Jang
- AMIST, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Hyunjin Kim
- Department of Neurology, Asan Medical Center, University of Ulsan, Seoul, 05505, South Korea
| | - Dong-Wha Kang
- Department of Neurology, Asan Medical Center, University of Ulsan, Seoul, 05505, South Korea
| | - Young-Min Lim
- Department of Neurology, Asan Medical Center, University of Ulsan, Seoul, 05505, South Korea
| | - Jaesoon Choi
- Biomedical Engineering, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| | - Eun-Jae Lee
- AMIST, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
- Department of Neurology, Asan Medical Center, University of Ulsan, Seoul, 05505, South Korea.
- Translational Biomedical Research Group, Asan Medical Center, University of Ulsan, Seoul, 05505, South Korea.
| |
Collapse
|
7
|
Hasaniani N, Nouri S, Shirzad M, Rostami-Mansoor S. Potential therapeutic and diagnostic approaches of exosomes in multiple sclerosis pathophysiology. Life Sci 2024; 347:122668. [PMID: 38670451 DOI: 10.1016/j.lfs.2024.122668] [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: 01/30/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
Exosomes are bilayer lipid vesicles that are released by cells and contain proteins, nucleic acids, and lipids. They can be internalized by other cells, inducing inflammatory responses and instigating toxicities in the recipient cells. Exosomes can also serve as therapeutic vehicles by transporting protective cargo to maintain homeostasis. Multiple studies have shown that exosomes can initiate and participate in the regulation of neuroinflammation, improve neurogenesis, and are closely related to the pathogenesis of central nervous system (CNS) diseases, including multiple sclerosis (MS). Exosomes can be secreted by both neurons and glial cells in the CNS, and their contents change with disease occurrence. Due to their ability to penetrate the blood-brain barrier and their stability in peripheral fluids, exosomes are attractive biomarkers of CNS diseases. In recent years, exosomes have emerged as potential therapeutic agents for CNS diseases, including MS. However, the molecular pathways in the pathogenesis of MS are still unknown, and further research is needed to fully understand the role of exosomes in the occurrence or improvement of MS disease. Thereby, in this review, we intend to provide a more complete understanding of the pathways in which exosomes are involved and affect the occurrence or improvement of MS disease.
Collapse
Affiliation(s)
- Nima Hasaniani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sina Nouri
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Urmia University of Medical Sciences, Urmia, Iran
| | - Moein Shirzad
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sahar Rostami-Mansoor
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
8
|
Schumacher SM, Doyle WJ, Hill K, Ochoa-Repáraz J. Gut microbiota in multiple sclerosis and animal models. FEBS J 2024:10.1111/febs.17161. [PMID: 38817090 PMCID: PMC11607183 DOI: 10.1111/febs.17161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/15/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024]
Abstract
Multiple sclerosis (MS) is a chronic central nervous system (CNS) neurodegenerative and neuroinflammatory disease marked by a host immune reaction that targets and destroys the neuronal myelin sheath. MS and correlating animal disease models show comorbidities, including intestinal barrier disruption and alterations of the commensal microbiome. It is accepted that diet plays a crucial role in shaping the microbiota composition and overall gastrointestinal (GI) tract health, suggesting an interplay between nutrition and neuroinflammation via the gut-brain axis. Unfortunately, poor host health and diet lead to microbiota modifications that could lead to significant responses in the host, including inflammation and neurobehavioral changes. Beneficial microbial metabolites are essential for host homeostasis and inflammation control. This review will highlight the importance of the gut microbiota in the context of host inflammatory responses in MS and MS animal models. Additionally, microbial community restoration and how it affects MS and GI barrier integrity will be discussed.
Collapse
Affiliation(s)
| | | | - Kristina Hill
- Department of Biological Sciences, Boise State University, Boise, ID 83725
| | | |
Collapse
|
9
|
Teekaput C, Thiankhaw K, Chattipakorn N, Chattipakorn SC. Possible Roles of Extracellular Vesicles in the Pathogenesis and Interventions of Immune-Mediated Central Demyelinating Diseases. Exp Neurobiol 2024; 33:47-67. [PMID: 38724476 PMCID: PMC11089403 DOI: 10.5607/en24002] [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: 01/30/2024] [Revised: 04/16/2024] [Accepted: 04/28/2024] [Indexed: 05/15/2024] Open
Abstract
Multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) are two of the most devastating immune-mediated central demyelinating disorders. NMOSD was once considered as a variant of MS until the discovery of an antibody specific to the condition. Despite both MS and NMOSD being considered central demyelinating disorders, their pathogenesis and clinical manifestations are distinct, however the exact mechanisms associated with each disease remain unclear. Extracellular vesicles (EVs) are nano-sized vesicles originating in various cells which serve as intercellular communicators. There is a large body of evidence to show the possible roles of EVs in the pathogenesis of several diseases, including the immune-mediated central demyelinating disorders. Various types of EVs are found across disease stages and could potentially be used as a surrogate marker, as well as acting by carrying a cargo of biochemical molecules. The possibility for EVs to be used as a next-generation targeted treatment for the immune-mediated central demyelinating disorders has been investigated. The aim of this review was to comprehensively identify, compile and discuss key findings from in vitro, in vivo and clinical studies. A summary of all findings shows that: 1) the EV profiles of MS and NMOSD differ from those of healthy individuals, 2) the use of EV markers as liquid biopsy diagnostic tools appears to be promising biomarkers for both MS and NMOSD, and 3) EVs are being studied as a potential targeted therapy for MS and NMOSD. Any controversial findings are also discussed in this review.
Collapse
Affiliation(s)
- Chutithep Teekaput
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kitti Thiankhaw
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
10
|
Sguigna PV, Hussain RZ, Okai A, Blackburn KM, Tardo L, Madinawala M, Korich J, Lebson LA, Kaplan J, Salter A, Manouchehri N, Stuve O. Cladribine tablets after treatment with natalizumab (CLADRINA) - rationale and design. Ther Adv Neurol Disord 2024; 17:17562864241233858. [PMID: 38585373 PMCID: PMC10996356 DOI: 10.1177/17562864241233858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/02/2024] [Indexed: 04/09/2024] Open
Abstract
Background Individual disease modifying therapies approved for multiple sclerosis (MS) have limited effectiveness and potentially serious side effects, especially when administered over long periods. Sequential combination therapy is a plausible alternative approach. Natalizumab is a monoclonal therapeutic antibody that reduces leukocyte access to the central nervous system that is associated with an increased risk of progressive multifocal leukoencephalopathy and disease reactivation after its discontinuation. Cladribine tablets act as a synthetic adenosine analog, disrupting DNA synthesis and repair, thereby reducing the number of lymphocytes. The generation of prospective, rigorous safety, and efficacy data in transitioning from natalizumab to cladribine is an unmet clinical need. Objectives To test the feasibility of transitioning patients with relapsing forms of MS natalizumab to cladribine tablets. Design Cladribine tablets after treatment with natalizumab (CLADRINA) is an open-label, single-arm, multicenter, collaborative phase IV, research study that will generate hypothesis regarding the safety, efficacy, and immunological impact of transition from natalizumab to cladribine tablets in patients with relapsing forms of MS. Methods and analysis Participants will be recruited from three different sites. The primary endpoint is the absolute and percent change from baseline of lymphocytes and myeloid cell subsets, as well as blood neurofilament light levels. The secondary endpoint is the annualized relapse rate over the 12- and 24-month trial periods. Exploratory endpoints include the expanded disability status scale, and magnetic resonance imaging outcomes. Discussion The CLADRINA trial will generate data regarding the safety, efficacy, and immunological impact of the transition from natalizumab to cladribine. As the pace of immunological knowledge of MS continues, insight into disease modifying therapy transition strategies is needed.
Collapse
Affiliation(s)
- Peter V. Sguigna
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rehana Z. Hussain
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Annette Okai
- North Texas Institute of Neurology & Headache, Plano, TX, USA
| | - Kyle M. Blackburn
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lauren Tardo
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mariam Madinawala
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Julie Korich
- EMD Serono, Inc., Rockland, MA, USA, an affiliate of Merck KGaA, Darmstadt, Germany
| | - Lori A. Lebson
- EMD Serono, Inc., Rockland, MA, USA, an affiliate of Merck KGaA, Darmstadt, Germany
| | - Jeffrey Kaplan
- Kansas City Multiple Sclerosis and Headache Center, Overland Park, KS, USA
| | - Amber Salter
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Navid Manouchehri
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390-8813, USA
- Neurology Section, VA North Texas Health Care System, Dallas, TX, USA
| |
Collapse
|
11
|
Kallal N, Hugues S, Garnier L. Regulation of autoimmune-mediated neuroinflammation by endothelial cells. Eur J Immunol 2024; 54:e2350482. [PMID: 38335316 DOI: 10.1002/eji.202350482] [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: 07/14/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
The CNS has traditionally been considered an immune-privileged organ, but recent studies have identified a plethora of immune cells in the choroid plexus, meninges, perivascular spaces, and cribriform plate. Although those immune cells are crucial for the maintenance of CNS homeostasis and for neural protection against infections, they can lead to neuroinflammation in some circumstances. The blood and the lymphatic vasculatures exhibit distinct structural and molecular features depending on their location in the CNS, greatly influencing the compartmentalization and the nature of CNS immune responses. In this review, we discuss how endothelial cells regulate the migration and the functions of T cells in the CNS both at steady-state and in murine models of neuroinflammation, with a special focus on the anatomical, cellular, and molecular mechanisms implicated in EAE.
Collapse
Affiliation(s)
- Neil Kallal
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Stephanie Hugues
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Laure Garnier
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
12
|
Kamyan D, Hassane M, Alnaqbi A, Souid AK, Rasbi ZA, Tahrawi AA, Shamsi MA. Ozanimod-mediated remission in experimental autoimmune encephalomyelitis is associated with enhanced activity of CNS CD27 low/- NK cell subset. Front Immunol 2024; 15:1230735. [PMID: 38533505 PMCID: PMC10963535 DOI: 10.3389/fimmu.2024.1230735] [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: 06/01/2023] [Accepted: 01/30/2024] [Indexed: 03/28/2024] Open
Abstract
Background Ozanimod (RPC1063) is an immunomodulator that has been recently approved by the FDA (2020) for the treatment of relapsing-remitting multiple sclerosis (RRMS). It is a selective agonist of the sphingosine-1-phophate receptors 1 and 5, expressed on naïve and central memory T and B cells, as well as natural killer (NK) cells, and is involved in lymphocyte trafficking. Oral administration of ozanimod was reported to result in rapid and reversible reduction in circulating lymphocytes in multiple sclerosis (MS) patients, however, only minimal effect on NK cells was observed. In this study, we sought to investigate the effect of ozanimod on NK cells and assess whether they play any role in ozanimod-induced remission in experimental autoimmune encephalomyelitis (EAE), the animal model of MS. Methods Active EAE induction was done in C57BL/6 female mice, followed by daily oral treatment with ozanimod (0.6mg/kg) starting at disease onset (score 1). Flow cytometry of blood and CNS was performed 24 hours after the last oral dose of ozanimod treatment in diseased mice. Histological analysis of lumbar spinal cord was performed for evaluating the level of inflammation and demyelination. Depletion of peripheral NK cells was done using anti-NK1.1 mouse antibody (mAb) at day 5 post-EAE induction. Results Ozanimod was effective in reducing the clinical severity of EAE and reducing the percentage of autoreactive CD4+ and CD8+ T cells along with significant inhibition of lymphocyte infiltration into the spinal cord, accompanied by reversed demyelination. Furthermore, ozanimod treatment resulted in a significant increase in the frequency of total NK cells in the blood and CNS along with upregulation of the activating receptor NKG2D on CD27low/- NK cell subset in the CNS. The effectiveness of ozanimod treatment in inhibiting the progression of the disease was reduced when NK cells were depleted using anti-NK1.1 mAb. Conclusion The current study demonstrated that ozanimod treatment significantly improved clinical symptoms in EAE mice. Ozanimod and anti-NK1.1 mAb appear to function in opposition to one another. Collectively, our data suggest that ozanimod-mediated remission is associated with an increased percentage of total NK cells and CD27low/- NK cells expressing the activating receptor, NKG2D in the CNS.
Collapse
Affiliation(s)
- Doua Kamyan
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Maya Hassane
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Alanood Alnaqbi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Abdul-Kader Souid
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Zakeya Al Rasbi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Abeer Al Tahrawi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Mariam Al Shamsi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
13
|
Tanaka Y, Ohki I, Murakami K, Ozawa S, Wang Y, Murakami M. The gateway reflex regulates tissue-specific autoimmune diseases. Inflamm Regen 2024; 44:12. [PMID: 38449060 PMCID: PMC10919025 DOI: 10.1186/s41232-024-00325-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/24/2024] [Indexed: 03/08/2024] Open
Abstract
The dynamic interaction and movement of substances and cells between the central nervous system (CNS) and peripheral organs are meticulously controlled by a specialized vascular structure, the blood-brain barrier (BBB). Experimental and clinical research has shown that disruptions in the BBB are characteristic of various neuroinflammatory disorders, including multiple sclerosis. We have been elucidating a mechanism termed the "gateway reflex" that details the entry of immune cells, notably autoreactive T cells, into the CNS at the onset of such diseases. This process is initiated through local neural responses to a range of environmental stimuli, such as gravity, electricity, pain, stress, light, and joint inflammation. These stimuli specifically activate neural pathways to open gateways at targeted blood vessels for blood immune cell entry. The gateway reflex is pivotal in managing tissue-specific inflammatory diseases, and its improper activation is linked to disease progression. In this review, we present a comprehensive examination of the gateway reflex mechanism.
Collapse
Affiliation(s)
- Yuki Tanaka
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
- Quantumimmunology Team, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan.
| | - Izuru Ohki
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Quantumimmunology Team, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kaoru Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Ozawa
- Quantumimmunology Team, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yaze Wang
- Quantumimmunology Team, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
- Quantumimmunology Team, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan.
- Division of Molecular Neuroimmunology, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan.
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan.
| |
Collapse
|
14
|
Li N, Han X, Ruan M, Huang F, Yang L, Xu T, Wang H, Wu H, Shi S, Wang Y, Wu X, Wang S. Prebiotic inulin controls Th17 cells mediated central nervous system autoimmunity through modulating the gut microbiota and short chain fatty acids. Gut Microbes 2024; 16:2402547. [PMID: 39287045 PMCID: PMC11409507 DOI: 10.1080/19490976.2024.2402547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory demyelination occurring in the central nervous system (CNS). Inulin is a common prebiotic that can improve metabolic disorders by modulating the gut microbiota. However, its capacity to affect CNS autoimmunity is poorly recognized. Experimental autoimmune encephalomyelitis (EAE) is a classical mouse model of MS. Herein, we found that oral administration of inulin ameliorated the severity EAE in mice, accompanied by reductions in inflammatory cell infiltration and demyelination in the CNS. These reductions were associated with decreased proportion and numbers of Th17 cells in brain and spleen. Consistent with the findings, the serum concentrations of IL-17, IL-6, and TNF-α were reduced in inulin treated EAE mice. Moreover, the proliferation of auto-reactive lymphocytes, against MOG35-55 antigen, was attenuated ex vivo. Mechanistically, inulin treatment altered the composition of gut microbiota. It increased Lactobacillus and Dubosiella whereas decreased g_Prevotellaceae_NK3B31_group at the genus level, alongside with elevated concentration of butyric acid in fecal content and serum. In vitro, butyrate, but not inulin, could inhibit the activation of MOG35-55 stimulated lymphocytes. Furthermore, fecal microbiota transplantation assay confirmed that fecal contents of inulin-treated normal mice had an ameliorative effect on EAE mice. In contrast, antibiotic cocktail (ABX) treatment diminished the therapeutic effect of inulin in EAE mice as well as the reduction of Th17 cells, while supplementation with Lactobacillus reuteri restored the amelioration effect. These results confirmed that the attenuation of inulin on Th17 cells and inflammatory demyelination in EAE mice was dependent on its modulation on gut microbiota and metabolites. Our findings provide a potential therapeutic regimen for prebiotic inulin supplementation in patients with multiple sclerosis.
Collapse
Affiliation(s)
- Ning Li
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyan Han
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming Ruan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Yang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianhao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huijun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Songshan Shi
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojun Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shunchun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Li M, Liu Q. Inflammatory Demyelinating Diseases of the Central Nervous System. ADVANCES IN NEUROBIOLOGY 2024; 41:171-218. [PMID: 39589715 DOI: 10.1007/978-3-031-69188-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Over the past decades, a large number of immunomodulatory or immunosuppressive treatments have been approved to treat central nervous system (CNS) demyelinating disorders such as multiple sclerosis (MS). Owing to the heterogeneity of patients with CNS demyelinating diseases, there is no clinical treatment that can adequately control all disease subtypes. Although significant progress has been made for relapsing-remitting MS, effective management of the progressive phase of MS has not yet been achieved. This is at least in part caused by our incomplete understanding of the mechanisms driving disease progression, despite our increasing knowledge regarding the underlying cellular and molecular mechanisms. Here, we summarized our current knowledge regarding the mechanisms of CNS demyelinating disorders and their animal models to identify open questions and challenges for existing concepts. We also discussed potential strategies for the future design of immune therapies to treat CNS demyelinating disorders.
Collapse
Affiliation(s)
- Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
16
|
Tanaka H, Hasebe R, Murakami K, Sugawara T, Yamasaki T, Murakami M. Gateway reflexes describe novel neuro-immune communications that establish immune cell gateways at specific vessels. Bioelectron Med 2023; 9:24. [PMID: 37936169 PMCID: PMC10631009 DOI: 10.1186/s42234-023-00126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023] Open
Abstract
Neuroinflammation is an important biological process induced by complex interactions between immune cells and neuronal cells in the central nervous system (CNS). Recent research on the bidirectional communication between neuronal and immunological systems has provided evidence for how immune and inflammatory processes are regulated by nerve activation. One example is the gateway reflex, in which immune cells bypass the blood brain barrier and infiltrate the CNS to cause neuroinflammation. We have found several modes of the gateway reflex in mouse models, in which gateways for immune cells are established at specific blood vessels in the spinal cords and brain in experimental autoimmune encephalomyelitis and systemic lupus erythematosus models, at retinal blood vessels in an experimental autoimmune uveitis model, and the ankle joints in an inflammatory arthritis model. Several environmental stimulations, including physical and psychological stresses, activate neurological pathways that alter immunological responses via the gateway reflex, thus contributing to the development/suppression of autoimmune diseases. In the manuscript, we describe the discovery of the gateway reflex and recent insights on how they regulate disease development. We hypothesize that artificial manipulation of specific neural pathways can establish and/or close the gateways to control the development of autoimmune diseases.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan.
| | - Rie Hasebe
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, national Institute for Natural Sciences, Nishi-38, Myodaiji-cho, Okazaki, 444-8585, Japan
| | - Kaoru Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
| | - Toshiki Sugawara
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
| | - Takeshi Yamasaki
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, national Institute for Natural Sciences, Nishi-38, Myodaiji-cho, Okazaki, 444-8585, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan.
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, national Institute for Natural Sciences, Nishi-38, Myodaiji-cho, Okazaki, 444-8585, Japan.
- Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Anagawa 4-9-1, Inage-Ku, Chiba, 263-8555, Japan.
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Nishi-11, Kita-21, Kuta-Ku, Sapporo, 001-0020, Japan.
| |
Collapse
|
17
|
Zhang W, Joshi C, Smith C, Ujas TA, Rivas JR, Cowell L, Christley S, Stowe AM, Monson NL. Neuronal binding by antibodies can be influenced by low pH stress during the isolation procedure. J Immunol Methods 2023; 521:113535. [PMID: 37558123 PMCID: PMC11249026 DOI: 10.1016/j.jim.2023.113535] [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: 01/05/2023] [Revised: 07/18/2023] [Accepted: 08/05/2023] [Indexed: 08/11/2023]
Abstract
Low pH stress and its influence on antibody binding is a common consideration among chemists, but is only recently emerging as a consideration in Immunological studies. Antibody characterizations in Multiple Sclerosis (MS), an autoimmune disease of the Central Nervous System (CNS) has revealed that antibodies in the cerebrospinal fluid (CSF) of patients with Multiple Sclerosis bind to myelin-related and non-myelin antigen targets. Many laboratories have used molecular biology techniques to generate recombinant human antibodies (rhAbs) expressed by individual B cells from healthy donors and patients with systemic autoimmune disease to identify antigen targets. This approach has been adapted within the Neuroimmunology research community to investigate antigen targets of individual B cells in the CSF of MS patients. Our laboratory determines which antibodies to clone based on their immunogenetics and this method enriches for cloning of rhAbs that bind to neurons. However, newer technologies to assist in purification of these rhAbs from culture supernatants use an acidic elution buffer which may enhance low pH stress on the antibody structure. Our laboratory routinely uses a basic elution buffer to purify rhAbs from culture supernatants to avoid low pH stress to the antibody structure. Our goal was to investigate whether acidic elution of our rhAbs using Next Generation Chromatography would impact the rhAbs' ability to bind neurons. The limited data presented here for two neuron-binding rhAbs tested indicated that acidic elution buffers used during rhAb purification impacted the ability of rhAbs with low CDR3 charge to maintain binding to neuronal targets. Reproducibility in a larger panel of rhAbs and factors underlying these observations remain untested.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Chaitanya Joshi
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Chad Smith
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Thomas A Ujas
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Jacqueline R Rivas
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Lindsay Cowell
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Scott Christley
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Ann M Stowe
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Nancy L Monson
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America.
| |
Collapse
|
18
|
Kendirli A, de la Rosa C, Lämmle KF, Eglseer K, Bauer IJ, Kavaka V, Winklmeier S, Zhuo L, Wichmann C, Gerdes LA, Kümpfel T, Dornmair K, Beltrán E, Kerschensteiner M, Kawakami N. A genome-wide in vivo CRISPR screen identifies essential regulators of T cell migration to the CNS in a multiple sclerosis model. Nat Neurosci 2023; 26:1713-1725. [PMID: 37709997 PMCID: PMC10545543 DOI: 10.1038/s41593-023-01432-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Multiple sclerosis (MS) involves the infiltration of autoreactive T cells into the CNS, yet we lack a comprehensive understanding of the signaling pathways that regulate this process. Here, we conducted a genome-wide in vivo CRISPR screen in a rat MS model and identified 5 essential brakes and 18 essential facilitators of T cell migration to the CNS. While the transcription factor ETS1 limits entry to the CNS by controlling T cell responsiveness, three functional modules, centered around the adhesion molecule α4-integrin, the chemokine receptor CXCR3 and the GRK2 kinase, are required for CNS migration of autoreactive CD4+ T cells. Single-cell analysis of T cells from individuals with MS confirmed that the expression of these essential regulators correlates with the propensity of CD4+ T cells to reach the CNS. Our data thus reveal key regulators of the fundamental step in the induction of MS lesions.
Collapse
Affiliation(s)
- Arek Kendirli
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Clara de la Rosa
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katrin F Lämmle
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Klara Eglseer
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Isabel J Bauer
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Vladyslav Kavaka
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Stephan Winklmeier
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - La Zhuo
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Christian Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Naoto Kawakami
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany.
| |
Collapse
|
19
|
Fitzsimons S, Muñoz-San Martín M, Nally F, Dillon E, Fashina IA, Strowitzki MJ, Ramió-Torrentà L, Dowling JK, De Santi C, McCoy CE. Inhibition of pro-inflammatory signaling in human primary macrophages by enhancing arginase-2 via target site blockers. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:941-959. [PMID: 37701067 PMCID: PMC10494319 DOI: 10.1016/j.omtn.2023.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 08/17/2023] [Indexed: 09/14/2023]
Abstract
The modulation of macrophage phenotype from a pro-inflammatory to an anti-inflammatory state holds therapeutic potential in the treatment of inflammatory disease. We have previously shown that arginase-2 (Arg2), a mitochondrial enzyme, is a key regulator of the macrophage anti-inflammatory response. Here, we investigate the therapeutic potential of Arg2 enhancement via target site blockers (TSBs) in human macrophages. TSBs are locked nucleic acid antisense oligonucleotides that were specifically designed to protect specific microRNA recognition elements (MREs) in human ARG2 3' UTR mRNA. TSBs targeting miR-155 (TSB-155) and miR-3202 (TSB-3202) MREs increased ARG2 expression in human monocyte-derived macrophages. This resulted in decreased gene expression and cytokine production of TNF-α and CCL2 and, for TSB-3202, in an increase in the anti-inflammatory macrophage marker, CD206. Proteomic analysis demonstrated that a network of pro-inflammatory responsive proteins was modulated by TSBs. In silico bioinformatic analysis predicted that TSB-3202 suppressed upstream pro-inflammatory regulators including STAT-1 while enhancing anti-inflammatory associated proteins. Proteomic data were validated by confirming increased levels of sequestosome-1 and decreased levels of phosphorylated STAT-1 and STAT-1 upon TSB treatment. In conclusion, upregulation of Arg2 by TSBs inhibits pro-inflammatory signaling and is a promising novel therapeutic strategy to modulate inflammatory signaling in human macrophages.
Collapse
Affiliation(s)
- Stephen Fitzsimons
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
- FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green, Dublin 2, Ireland
| | - María Muñoz-San Martín
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Frances Nally
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Eugene Dillon
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ifeolutembi A. Fashina
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Moritz J. Strowitzki
- Department of General, Visceral & Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Lluís Ramió-Torrentà
- Neuroinflammation and Neurodegeneration Group, Girona Biomedical Research Institute (IDIBGI), CERCA Programme/Generalitat de Catalunya, Salt, Girona, Spain
| | - Jennifer K. Dowling
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
- FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green, Dublin 2, Ireland
| | - Chiara De Santi
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Claire E. McCoy
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
- FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green, Dublin 2, Ireland
| |
Collapse
|
20
|
Bechara R, Vagner S, Mariette X. Post-transcriptional checkpoints in autoimmunity. Nat Rev Rheumatol 2023; 19:486-502. [PMID: 37311941 DOI: 10.1038/s41584-023-00980-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 06/15/2023]
Abstract
Post-transcriptional regulation is a fundamental process in gene expression that has a role in diverse cellular processes, including immune responses. A core concept underlying post-transcriptional regulation is that protein abundance is not solely determined by transcript abundance. Indeed, transcription and translation are not directly coupled, and intervening steps occur between these processes, including the regulation of mRNA stability, localization and alternative splicing, which can impact protein abundance. These steps are controlled by various post-transcription factors such as RNA-binding proteins and non-coding RNAs, including microRNAs, and aberrant post-transcriptional regulation has been implicated in various pathological conditions. Indeed, studies on the pathogenesis of autoimmune and inflammatory diseases have identified various post-transcription factors as important regulators of immune cell-mediated and target effector cell-mediated pathological conditions. This Review summarizes current knowledge regarding the roles of post-transcriptional checkpoints in autoimmunity, as evidenced by studies in both haematopoietic and non-haematopoietic cells, and discusses the relevance of these findings for developing new anti-inflammatory therapies.
Collapse
Affiliation(s)
- Rami Bechara
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Le Kremlin Bicêtre, France.
| | - Stephan Vagner
- Institut Curie, CNRS UMR3348, INSERM U1278, PSL Research University, Université Paris-Saclay, Orsay, France
| | - Xavier Mariette
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Le Kremlin Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Hôpital Bicêtre, Department of Rheumatology, Le Kremlin Bicêtre, France
| |
Collapse
|
21
|
Fan J, Han Y, Sun H, Sun S, Wang Y, Guo R, Guo J, Tian X, Wang J, Wang J. Mesenchymal stem cell-derived exosomal microRNA-367–3p alleviates experimental autoimmune encephalomyelitis via inhibition of microglial ferroptosis by targeting EZH2. Biomed Pharmacother 2023; 162:114593. [PMID: 37001184 DOI: 10.1016/j.biopha.2023.114593] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/11/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune, inflammatory demyelinating disorder of the central nervous system. Accumulating evidence has underscored the therapeutic potential of bone marrow mesenchymal stem cells (BMSCs)-derived exosomes (BMSC-Exos) containing bioactive compounds in MS. Herein, the current study sought to characterize the mechanism of BMSC-Exos harboring miR-367-3p both in BV2 microglia by Erastin-induced ferroptosis and in experimental autoimmune encephalomyelitis (EAE), a typical animal model of MS. Exosomes were firstly isolated from BMSCs and identified for further use. BV2 microglia were co-cultured with miR-367-3p-containing BMSC-Exos, followed by an assessment of cell ferroptosis. Mechanistic exploration was furthered by the interaction of miR-367-3p and its downstream regulators. Lastly, BMSC-Exos harboring miR-367-3p were injected into EAE mice for in vivo validation. BMSC-Exos carrying miR-367-3p restrained microglial ferroptosis in vitro. Mechanistically, miR-367-3p could bind to Enhancer of zeste homolog 2 (EZH2) and restrain EZH2 expression, leading to the over-expression of solute carrier family 7 member 11 (SLC7A11). Meanwhile, over-expression of SLC7A11 resulted in Glutathione Peroxidase 4 (GPX4) activation and ferroptosis suppression. Ectopic expression of EZH2 in vitro negated the protective effects of BMSC-Exos. Furthermore, BMSC-Exos containing miR-367-3p relieved the severity of EAE by suppressing ferroptosis and restraining EZH2 expression in vivo. Collectively, our findings suggest that BMSC-Exos carrying miR-367-3p brings about a significant decline in microglia ferroptosis by repressing EZH2 and alleviating the severity of EAE in vivo, suggesting a possible role of miR-367-3p overexpression in the treatment strategy of EAE. AVAILABILITY OF DATA AND MATERIALS: The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.
Collapse
|
22
|
Li Z, Antila S, Nurmi H, Chilov D, Korhonen EA, Fang S, Karaman S, Engelhardt B, Alitalo K. Blockade of VEGFR3 signaling leads to functional impairment of dural lymphatic vessels without affecting autoimmune neuroinflammation. Sci Immunol 2023; 8:eabq0375. [PMID: 37058549 DOI: 10.1126/sciimmunol.abq0375] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
The recent discovery of lymphatic vessels (LVs) in the dura mater, the outermost layer of meninges around the central nervous system (CNS), has opened a possibility for the development of alternative therapeutics for CNS disorders. The vascular endothelial growth factor C (VEGF-C)/VEGF receptor 3 (VEGFR3) signaling pathway is essential for the development and maintenance of dural LVs. However, its significance in mediating dural lymphatic function in CNS autoimmunity is unclear. We show that inhibition of the VEGF-C/VEGFR3 signaling pathway using a monoclonal VEGFR3-blocking antibody, a soluble VEGF-C/D trap, or deletion of the Vegfr3 gene in adult lymphatic endothelium causes notable regression and functional impairment of dural LVs but has no effect on the development of CNS autoimmunity in mice. During autoimmune neuroinflammation, the dura mater was only minimally affected, and neuroinflammation-induced helper T (TH) cell recruitment, activation, and polarization were significantly less pronounced in the dura mater than in the CNS. In support of this notion, during autoimmune neuroinflammation, blood vascular endothelial cells in the cranial and spinal dura expressed lower levels of cell adhesion molecules and chemokines, and antigen-presenting cells (i.e., macrophages and dendritic cells) had lower expression of chemokines, MHC class II-associated molecules, and costimulatory molecules than their counterparts in the brain and spinal cord, respectively. The significantly weaker TH cell responses in the dura mater may explain why dural LVs do not contribute directly to CNS autoimmunity.
Collapse
Affiliation(s)
- Zhilin Li
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Salli Antila
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Harri Nurmi
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Dmitri Chilov
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Emilia A Korhonen
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Shentong Fang
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Sinem Karaman
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | | | - Kari Alitalo
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| |
Collapse
|
23
|
Novakova L, Henricsson M, Björnson E, Axelsson M, Borén J, Rosenstein I, Lycke J, Cardell SL, Blomqvist M. Cerebrospinal fluid sulfatide isoforms lack diagnostic utility in separating progressive from relapsing-remitting multiple sclerosis. Mult Scler Relat Disord 2023; 74:104705. [PMID: 37060853 DOI: 10.1016/j.msard.2023.104705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 04/02/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is an immune-mediated demyelinating disorder of the central nervous system. The glycosphingolipid sulfatide, a lipid particularly enriched in the myelin sheath, has been shown to be involved the maintenance of this specific membrane structure. Sulfatide in cerebrospinal fluid (CSF) may reflect demyelination, a dominating feature of MS. We investigated the diagnostic utility of CSF sulfatide isoform levels to separate different courses or phenotypes of MS disease. MATERIAL AND METHODS This was a mono-center, cross-sectional study of relapsing-remitting MS (RRMS) (n = 45) and progressive MS (PMS) (n = 42) patients (consisting of primary PMS (n = 17) and secondary PMS (n = 25)) and healthy controls (n = 19). In total, 20 sulfatide isoforms were measured in CSF by liquid chromatography-mass spectrometry. RESULTS CSF total sulfatide concentrations, as well as CSF sulfatide isoform distribution, did not differ across the study groups, and their levels were independent of disease course/phenotype, disease duration, time to conversion to secondary PMS, age, and disability in MS patients. CONCLUSION CSF sulfatide isoforms lack diagnostic and prognostic utility as a biomarker for progressive MS.
Collapse
Affiliation(s)
- Lenka Novakova
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine/Wallenberg Lab, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Elias Björnson
- Department of Molecular and Clinical Medicine/Wallenberg Lab, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Markus Axelsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Lab, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Igal Rosenstein
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susanna L Cardell
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria Blomqvist
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden; Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg 413 85, Sweden.
| |
Collapse
|
24
|
Wang J, Sun H, Guo R, Guo J, Tian X, Wang J, Sun S, Han Y, Wang Y. Exosomal miR-23b-3p from bone mesenchymal stem cells alleviates experimental autoimmune encephalomyelitis by inhibiting microglial pyroptosis. Exp Neurol 2023; 363:114374. [PMID: 36907352 DOI: 10.1016/j.expneurol.2023.114374] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/18/2023] [Accepted: 03/02/2023] [Indexed: 03/14/2023]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease that affects the central nervous system and is marked by inflammation and damage to the myelin sheath surrounding nerve fibers. Recent studies have highlighted the therapeutic value of exosomes (Exos) obtained from bone marrow mesenchymal stem cells (BMSCs) in MS treatment. These BMSC-Exos contain biologically active molecules that show promising results in preclinical evaluations. The aim of this study was to investigate the mechanism of BMSC-Exos containing miR-23b-3p in both LPS-stimulated BV2 microglia and in experimental autoimmune encephalomyelitis (EAE), an animal model for MS. Exos were isolated from BMSCs, and their effects were evaluated in vitro by co-culturing with BV2 microglia. The interaction between miR-23b-3p and its downstream targets was also explored. The efficacy of BMSC-Exos was further verified in vivo by injecting the Exos into EAE mice. The results showed that BMSC-Exos containing miR-23b-3p reduced microglial pyroptosis in vivo by specifically binding to and suppressing the expression of NEK7. In vivo, BMSC-Exos containing miR-23b-3p alleviated the severity of EAE by decreasing microglial inflammation and pyroptosis via the repression of NEK7. These findings provide new insights into the therapeutic potential of BMSC-Exos containing miR-23b-3p for MS.
Collapse
Affiliation(s)
- Jueqiong Wang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huanhuan Sun
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ruoyi Guo
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiangyuan Guo
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Xinyi Tian
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jinli Wang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shichao Sun
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yusen Han
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ying Wang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
25
|
Li J, Lu L, Binder K, Xiong J, Ye L, Cheng YH, Majri-Morrison S, Lu W, Lee JW, Zhang Z, Wu YZ, Zheng L, Lenardo MJ. Mechanisms of antigen-induced reversal of CNS inflammation in experimental demyelinating disease. SCIENCE ADVANCES 2023; 9:eabo2810. [PMID: 36857453 PMCID: PMC9977187 DOI: 10.1126/sciadv.abo2810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 12/27/2022] [Indexed: 06/18/2023]
Abstract
Autoimmune central nervous system (CNS) demyelinating diseases are a major public health burden and poorly controlled by current immunosuppressants. More precise immunotherapies with higher efficacy and fewer side effects are sought. We investigated the effectiveness and mechanism of an injectable myelin-based antigenic polyprotein MMPt (myelin oligodendrocyte glycoprotein, myelin basic protein and proteolipid protein, truncated). We find that it suppresses mouse experimental autoimmune encephalomyelitis without major side effects. MMPt induces rapid apoptosis of the encephalitogenic T cells and suppresses inflammation in the affected CNS. Intravital microscopy shows that MMPt is taken up by perivascular F4/80+ cells but not conventional antigen-presenting dendritic cells, B cells, or microglia. MMPt-stimulated F4/80+ cells induce reactive T cell immobilization and apoptosis in situ, resulting in reduced infiltration of inflammatory cells and chemokine production. Our study reveals alternative mechanisms that explain how cognate antigen suppresses CNS inflammation and may be applicable for effectively and safely treating demyelinating diseases.
Collapse
Affiliation(s)
- Jian Li
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lisen Lu
- MoE Key Laboratory for Biomedical Photonics, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Kyle Binder
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jian Xiong
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan H. Cheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sonia Majri-Morrison
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wei Lu
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jae W. Lee
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zhihong Zhang
- MoE Key Laboratory for Biomedical Photonics, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yu-zhang Wu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lixin Zheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael J. Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Charabati M, Wheeler MA, Weiner HL, Quintana FJ. Multiple sclerosis: Neuroimmune crosstalk and therapeutic targeting. Cell 2023; 186:1309-1327. [PMID: 37001498 PMCID: PMC10119687 DOI: 10.1016/j.cell.2023.03.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/23/2023] [Accepted: 03/03/2023] [Indexed: 04/03/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of the central nervous system afflicting nearly three million individuals worldwide. Neuroimmune interactions between glial, neural, and immune cells play important roles in MS pathology and offer potential targets for therapeutic intervention. Here, we review underlying risk factors, mechanisms of MS pathogenesis, available disease modifying therapies, and examine the value of emerging technologies, which may address unmet clinical needs and identify novel therapeutic targets.
Collapse
Affiliation(s)
- Marc Charabati
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
27
|
Wu N, Li X, Ma H, Zhang X, Liu B, Wang Y, Zheng Q, Fan X. The role of the gut microbiota and fecal microbiota transplantation in neuroimmune diseases. Front Neurol 2023; 14:1108738. [PMID: 36816570 PMCID: PMC9929158 DOI: 10.3389/fneur.2023.1108738] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
The gut microbiota plays a key role in the function of the host immune system and neuroimmune diseases. Alterations in the composition of the gut microbiota can lead to pathology and altered formation of microbiota-derived components and metabolites. A series of neuroimmune diseases, such as myasthenia gravis (MG), multiple sclerosis (MS), neuromyelitis optica spectrum disorders (NMOSDs), Guillain-Barré syndrome (GBS), and autoimmune encephalitis (AIE), are associated with changes in the gut microbiota. Microecological therapy by improving the gut microbiota is expected to be an effective measure for treating and preventing some neuroimmune diseases. This article reviews the research progress related to the roles of gut microbiota and fecal microbiota transplantation (FMT) in neuroimmune diseases.
Collapse
Affiliation(s)
- Nan Wu
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Xizhi Li
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - He Ma
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Xue Zhang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Bin Liu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Yuan Wang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China,*Correspondence: Yuan Wang ✉
| | - Qi Zheng
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China,Qi Zheng ✉
| | - Xueli Fan
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China,Xueli Fan ✉
| |
Collapse
|
28
|
Dericioğlu V, Akkaya Turhan S, Erdem HE, Sevik MO, Erdil E, Sünter G, Ağan K, Toker E. In Vivo Corneal Confocal Microscopy in Multiple Sclerosis: Can it Differentiate Disease Relapse in Multiple Sclerosis? Am J Ophthalmol 2023; 250:138-148. [PMID: 36669610 DOI: 10.1016/j.ajo.2023.01.015] [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: 07/12/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023]
Abstract
PURPOSE This study aims to investigate the role of in vivo corneal confocal microscopy (IVCCM) in the detection of corneal inflammatory activity and subbasal nerve alterations in patients with multiple sclerosis (MS) and to further determine whether IVCCM can be used to detect (acute) disease relapse. DESIGN Prospective cross-sectional study, with a subgroup follow-up. METHODS This single-center study included 58 patients with MS (MS-Relapse group [n = 27] and MS-Remission group [n = 31]), and 30 age- and sex-matched healthy control subjects. Patients with a history of optic neuritis or trigeminal symptoms were excluded. Corneal nerve fiber density (CNFD), corneal nerve branch density (CNBD), corneal nerve fiber length (CNFL), and dendritic cell (DC) density were evaluated in all patients with MS and control subjects by IVCCM. Patients in the MS-Relapse group who were in remission for ≥6 months after the MS incident underwent a repeat IVCCM. RESULTS No statistical difference was observed between the MS-Relapse and MS-Remission groups regarding age, sex, MS duration, and the number of relapses (P > .05). Compared with healthy control subjects, all subbasal nerve parameters were significantly lower (CNFD: P < .001, CNFL: P < .001, CNBD: P < .001), and the DC density was significantly higher (P = .023) in patients with MS. However, no significant difference was observed between MS-Relapse and MS-Remission groups in terms of CNFD (mean [SE] difference -2.05 [1.69] fibers/mm2 [95% confidence interval {CI} -1.32 to 5.43]; P < .227), CNFL (mean [SE] difference -1.10 [0.83] mm/mm2 [95% CI -0.56 to 2.75]; P < .190), CNBD (mean [SE] difference -3.91 [2.48] branches/mm2 [95% CI -1.05 to 8.87]; P < .120), and DC density (median [IQR], 59.38 [43.75-85.0] vs 75.0 [31.25-128.75]; P = .596). The repeat IVCCM in relapse patients (n = 16 [59.3%]) showed a significant increase in CNFD (P = .036) and CNBD (P = .018), but no change was observed in CNFL (P = .075) and DC density (P = .469). CONCLUSION Although increased inflammation and neurodegeneration can be demonstrated in patients with MS compared with healthy control subjects, a single time point evaluation of IVCCM does not seem to be sufficient to confirm the occurrence of relapse in patients with MS. However, IVCCM holds promise for demonstrating early neuroregeneration in patients with MS.
Collapse
Affiliation(s)
- Volkan Dericioğlu
- From the Department of Ophthalmology (V.D., S.A.T., H.E.E., M.O.S.), Marmara University School of Medicine, Istanbul, Turkey.
| | - Semra Akkaya Turhan
- From the Department of Ophthalmology (V.D., S.A.T., H.E.E., M.O.S.), Marmara University School of Medicine, Istanbul, Turkey
| | - Halit Eren Erdem
- From the Department of Ophthalmology (V.D., S.A.T., H.E.E., M.O.S.), Marmara University School of Medicine, Istanbul, Turkey
| | - Mehmet Orkun Sevik
- From the Department of Ophthalmology (V.D., S.A.T., H.E.E., M.O.S.), Marmara University School of Medicine, Istanbul, Turkey
| | - Esra Erdil
- and the Department of Neurology (E.E., G.S., K.A.), Marmara University School of Medicine, Istanbul, Turkey
| | - Gülin Sünter
- and the Department of Neurology (E.E., G.S., K.A.), Marmara University School of Medicine, Istanbul, Turkey
| | - Kadriye Ağan
- and the Department of Neurology (E.E., G.S., K.A.), Marmara University School of Medicine, Istanbul, Turkey
| | - Ebru Toker
- and the Department of Ophthalmology and Visual Sciences (E.T.), West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
29
|
Dendrimers in Neurodegenerative Diseases. Processes (Basel) 2023. [DOI: 10.3390/pr11020319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Neurodegenerative diseases (NDs), such as Parkinson’s Disease (PD), Alzheimer’s Disease (AD), Multiple Sclerosis (MS) and amyotrophic lateral sclerosis (ALS), are characterized by progressive loss of structure or function of neurons. Current therapies for NDs are only symptomatic and long-term ineffective. This challenge has promoted the development of new therapies against relevant targets in these pathologies. In this review, we will focus on the most promising therapeutic approaches based on dendrimers (DDs) specially designed for the treatment and diagnosis of NDs. DDs are well-defined polymeric structures that provide a multifunctional platform for developing different nanosystems for a myriad of applications. DDs have been proposed as interesting drug delivery systems with the ability to cross the blood–brain barrier (BBB) and increase the bioavailability of classical drugs in the brain, as well as genetic material, by reducing the synthesis of specific targets, as β-amyloid peptide. Moreover, DDs have been shown to be promising anti-amyloidogenic systems against amyloid-β peptide (Aβ) and Tau aggregation, powerful agents for blocking α-synuclein (α-syn) fibrillation, exhibit anti-inflammatory properties, promote cellular uptake to certain cell types, and are potential tools for ND diagnosis. In summary, DDs have emerged as promising alternatives to current ND therapies since they may limit the extent of damage and provide neuroprotection to the affected tissues.
Collapse
|
30
|
Jalkanen S, Salmi M. Lymphocyte Adhesion and Trafficking. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00016-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
31
|
Akabane K, Murakami K, Murakami M. Gateway reflexes are neural circuits that establish the gateway of immune cells to regulate tissue specific inflammation. Expert Opin Ther Targets 2023; 27:469-477. [PMID: 37318003 DOI: 10.1080/14728222.2023.2225215] [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: 12/07/2022] [Accepted: 06/11/2023] [Indexed: 06/16/2023]
Abstract
INTRODUCTION Tissue-specific inflammatory diseases are regulated by several mechanisms. The gateway reflex and IL-6 amplifier are two mechanisms involved in diseases that depend on the inflammatory cytokine IL-6. The gateway reflex activates specific neural pathways that cause autoreactive CD4+ T cells to pass through gateways in blood vessels toward specific tissues in tissue-specific inflammatory diseases. These gateways are mediated by the IL-6 amplifier, which describes enhanced NF-κB activation in nonimmune cells including endothelial cells at specific sites. In total, we have reported six gateway reflexes defined by their triggering stimulus: gravity, pain, electric stimulation, stress, light, and joint inflammation. AREAS COVERED This review summarizes the gateway reflex and IL-6 amplifier for the development of tissue-specific inflammatory diseases. EXPERT OPINION We expect that the IL-6 amplifier and gateway reflex will lead to novel therapeutic and diagnostic methods for inflammatory diseases, particularly tissue-specific ones.
Collapse
Affiliation(s)
- Keiichiroh Akabane
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kaoru 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
| | - 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 Molecular Neuroimmunology, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Institute for Vaccine Research and Development(HU-IVRed), Hokkaido University, Sapporo, Japan
| |
Collapse
|
32
|
Ineichen BV, Okar SV, Proulx ST, Engelhardt B, Lassmann H, Reich DS. Perivascular spaces and their role in neuroinflammation. Neuron 2022; 110:3566-3581. [PMID: 36327898 PMCID: PMC9905791 DOI: 10.1016/j.neuron.2022.10.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 08/17/2022] [Accepted: 10/13/2022] [Indexed: 11/19/2022]
Abstract
It is uncontested that perivascular spaces play critical roles in maintaining homeostasis and priming neuroinflammation. However, despite more than a century of intense research on perivascular spaces, many open questions remain about the anatomical compartment surrounding blood vessels within the CNS. The goal of this comprehensive review is to summarize the literature on perivascular spaces in human neuroinflammation and associated animal disease models. We describe the cell types taking part in the morphological and functional aspects of perivascular spaces and how those spaces can be visualized. Based on this, we propose a model of the cascade of events occurring during neuroinflammatory pathology. We also discuss current knowledge gaps and limitations of the available evidence. An improved understanding of perivascular spaces could advance our comprehension of the pathophysiology of neuroinflammation and open a new therapeutic window for neuroinflammatory diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Benjamin V Ineichen
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Center for Reproducible Science, University of Zurich, Zurich, Switzerland.
| | - Serhat V Okar
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven T Proulx
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
33
|
Tahedl M, Levine SM, Weissert R, Kohl Z, Lee DH, Linker RA, Schwarzbach JV. Early remission in multiple sclerosis is linked to altered coherence of the Cerebellar Network. J Transl Med 2022; 20:488. [PMID: 36303221 PMCID: PMC9615296 DOI: 10.1186/s12967-022-03576-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/06/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The development of permanent disability in multiple sclerosis (MS) is highly variable among patients, and the exact mechanisms that contribute to this disability remain unknown. METHODS Following the idea that the brain has intrinsic network organization, we investigated changes of functional networks in MS patients to identify possible links between network reorganization and remission from clinical episodes in MS. Eighteen relapsing-remitting MS patients (RRMS) in their first clinical manifestation underwent resting-state functional MRI and again during remission. We used ten template networks, identified from independent component analysis, to compare changes in network coherence for each patient compared to those of 44 healthy controls from the Human Connectome Project test-retest dataset (two-sample t-test of pre-post differences). Combining a binomial test with Monte Carlo procedures, we tested four models of how functional coherence might change between the first clinical episode and remission: a network can change its coherence (a) with itself ("one-with-self"), (b) with another network ("one-with-other"), or (c) with a set of other networks ("one-with-many"), or (d) multiple networks can change their coherence with respect to one common network ("many-with-one"). RESULTS We found evidence supporting two of these hypotheses: coherence decreased between the Executive Control Network and several other networks ("one-with-many" hypothesis), and a set of networks altered their coherence with the Cerebellar Network ("many-with-one" hypothesis). CONCLUSION Given the unexpected commonality of the Cerebellar Network's altered coherence with other networks (a finding present in more than 70% of the patients, despite their clinical heterogeneity), we conclude that remission in MS may result from learning processes mediated by the Cerebellar Network.
Collapse
Affiliation(s)
- Marlene Tahedl
- grid.7727.50000 0001 2190 5763Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany ,grid.7727.50000 0001 2190 5763Institute for Psychology, University of Regensburg, 93053 Regensburg, Germany
| | - Seth M. Levine
- grid.5252.00000 0004 1936 973XDepartment of Psychology, LMU Munich, 80802 Munich, Germany ,grid.411095.80000 0004 0477 2585NeuroImaging Core Unit Munich (NICUM), University Hospital LMU, 80336 Munich, Germany
| | - Robert Weissert
- grid.7727.50000 0001 2190 5763Department of Neurology, University of Regensburg, 93053 Regensburg, Germany
| | - Zacharias Kohl
- grid.7727.50000 0001 2190 5763Department of Neurology, University of Regensburg, 93053 Regensburg, Germany
| | - De-Hyung Lee
- grid.7727.50000 0001 2190 5763Department of Neurology, University of Regensburg, 93053 Regensburg, Germany
| | - Ralf A. Linker
- grid.7727.50000 0001 2190 5763Department of Neurology, University of Regensburg, 93053 Regensburg, Germany
| | - Jens V. Schwarzbach
- grid.7727.50000 0001 2190 5763Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
34
|
Zhu Y, Owens SJ, Murphy CE, Ajulu K, Rothmond D, Purves-Tyson T, Middleton F, Webster MJ, Weickert CS. Inflammation-related transcripts define "high" and "low" subgroups of individuals with schizophrenia and bipolar disorder in the midbrain. Brain Behav Immun 2022; 105:149-159. [PMID: 35764269 DOI: 10.1016/j.bbi.2022.06.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/10/2022] [Accepted: 06/23/2022] [Indexed: 01/08/2023] Open
Abstract
Dopamine dysregulation in schizophrenia may be associated with midbrain inflammation. Previously, we found elevated levels of pro-inflammatory cytokine mRNAs in the post-mortem midbrain of people with schizophrenia (46%) but not from unaffected controls (0%) using a brain cohort from Sydney, Australia. Here, we measured cytokine mRNAs and proteins in the midbrain in the Stanley Medical Research Institute (SMRI) array cohort (N = 105). We tested if the proportions of individuals with schizophrenia and with high inflammation can be replicated, and if individuals with bipolar disorder with elevated midbrain cytokines can be identified. mRNA levels of 7 immune transcripts from post-mortem midbrain tissue were measured via RT-PCR and two-step recursive clustering analysis was performed using 4 immune transcripts to define "high and low" inflammatory subgroups. The clustering predictors used were identical to our earlier midbrain study, and included: IL1B, IL6, TNF, and SERPINA3 mRNA levels. 46% of schizophrenia cases (16/35 SCZ), 6% of controls (2/33 CTRL), and 29% of bipolar disorder cases (10/35 BPD) were identified as belonging to the high inflammation (HI) subgroups [χ2 (2) = 13.54, p < 0.001]. When comparing inflammatory subgroups, all four mRNAs were significantly increased in SCZ-HI and BPD-HI compared to low inflammation controls (CTRL-LI) (p < 0.05). Additionally, protein levels of IL-1β, IL-6, and IL-18 were elevated in SCZ-HI and BPD-HI compared to all other low inflammatory subgroups (all p < 0.05). Surprisingly, TNF-α protein levels were unchanged according to subgroups. In conclusion, we determined that almost half of the individuals with schizophrenia were defined as having high inflammation in the midbrain, replicating our previous findings. Further, we detected close to one-third of those with bipolar disorder to be classified as having high inflammation. Elevations in some pro-inflammatory cytokine mRNAs (IL-1β and IL-6) were also found at the protein level, whereas TNF mRNA and protein levels were not concordant.
Collapse
Affiliation(s)
- Yunting Zhu
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Samantha J Owens
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Caitlin E Murphy
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Kachikwulu Ajulu
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Debora Rothmond
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Tertia Purves-Tyson
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Frank Middleton
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, 9800 Medical Center Drive, Rockville, MD, USA
| | - Cynthia Shannon Weickert
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY 13210, USA; Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW 2031, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
35
|
Zuroff L, Rezk A, Shinoda K, Espinoza DA, Elyahu Y, Zhang B, Chen AA, Shinohara RT, Jacobs D, Alcalay RN, Tropea TF, Chen-Plotkin A, Monsonego A, Li R, Bar-Or A. Immune aging in multiple sclerosis is characterized by abnormal CD4 T cell activation and increased frequencies of cytotoxic CD4 T cells with advancing age. EBioMedicine 2022; 82:104179. [PMID: 35868128 PMCID: PMC9305354 DOI: 10.1016/j.ebiom.2022.104179] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 06/12/2022] [Accepted: 07/05/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Immunosenescence (ISC) describes age-related changes in immune-system composition and function. Multiple sclerosis (MS) is a lifelong inflammatory condition involving effector and regulatory T-cell imbalance, yet little is known about T-cell ISC in MS. We examined age-associated changes in circulating T cells in MS compared to normal controls (NC). METHODS Forty untreated MS (Mean Age 43·3, Range 18-72) and 49 NC (Mean Age 48·6, Range 20-84) without inflammatory conditions were included in cross-sectional design. T-cell subsets were phenotypically and functionally characterized using validated multiparametric flow cytometry. Their aging trajectories, and differences between MS and NC, were determined using linear mixed-effects models. FINDINGS MS patients demonstrated early and persistent redistribution of naïve and memory CD4 T-cell compartments. While most CD4 and CD8 T-cell aging trajectories were similar between groups, MS patients exhibited abnormal age-associated increases of activated (HLA-DR+CD38+; (P = 0·013) and cytotoxic CD4 T cells, particularly in patients >60 (EOMES: P < 0·001). Aging MS patients also failed to upregulate CTLA-4 expression on both CD4 (P = 0·014) and CD8 (P = 0·009) T cells, coupled with abnormal age-associated increases in frequencies of B cells expressing costimulatory molecules. INTERPRETATION While many aspects of T-cell aging in MS are conserved, the older MS patients harbour abnormally increased frequencies of CD4 T cells with activated and cytotoxic effector profiles. Age-related decreased expression of T-cell co-inhibitory receptor CTLA-4, and increased B-cell costimulatory molecule expression, may provide a mechanism that drives aberrant activation of effector CD4 T cells that have been implicated in progressive disease. FUNDING Stated in Acknowledgements section of manuscript.
Collapse
Affiliation(s)
- Leah Zuroff
- The Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ayman Rezk
- The Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Koji Shinoda
- The Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Diego A Espinoza
- The Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yehezqel Elyahu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences; Zlotowski Neuroscience Center and Regenerative Medicine and Stem Cell Research Center; and National Institute for Biotechnology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Bo Zhang
- Department of Cardiology, The fourth affiliated hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Andrew A Chen
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology, and Informatics, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dina Jacobs
- The Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University, New York, NY 10032, USA; The Center for Movement Disorders, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6423914, Israel
| | - Thomas F Tropea
- Department of Neurology, Perelman school of medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman school of medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alon Monsonego
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences; Zlotowski Neuroscience Center and Regenerative Medicine and Stem Cell Research Center; and National Institute for Biotechnology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Rui Li
- The Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Amit Bar-Or
- The Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
36
|
Colciaghi F, Costanza M. Unveiling Leukocyte Extracellular Traps in Inflammatory Responses of the Central Nervous System. Front Immunol 2022; 13:915392. [PMID: 35844591 PMCID: PMC9283689 DOI: 10.3389/fimmu.2022.915392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Over the past nearly two decades, increasing evidence has uncovered how immune cells can actively extrude genetic material to entrap invading pathogens or convey sterile inflammatory signals that contribute to shaping immune responses. Originally identified in neutrophils, the release of decondensed chromatin fibers decorated with antimicrobial proteins, called extracellular traps (ETs), has been recognized as a specific form of programmed inflammatory cell death, which is now known to occur in several other leukocytes. Subsequent reports have shown that self-DNA can be extruded from immune cells even in the absence of cell death phenomena. More recent data suggest that ETs formation could exacerbate neuroinflammation in several disorders of the central nervous system (CNS). This review article provides an overview of the varied types, sources, and potential functions of extracellular DNA released by immune cells. Key evidence suggesting the involvement of ETs in neurodegenerative, traumatic, autoimmune, and oncological disorders of the CNS will be discussed, outlining ongoing challenges and drawing potentially novel lines of investigation.
Collapse
Affiliation(s)
- Francesca Colciaghi
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Massimo Costanza
- Molecular Neuro-Oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- *Correspondence: Massimo Costanza,
| |
Collapse
|
37
|
Caminade AM, Turrin CO, Poupot R. Curing inflammatory diseases using phosphorous dendrimers. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1783. [PMID: 35194953 DOI: 10.1002/wnan.1783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Different types of water-soluble phosphorous dendrimers have been synthesized and display many different biological properties. It has been shown in particular that phosphorous dendrimers of first generation functionalized with azabisphosphonate terminal functions are able to stimulate the human immune system ex vivo. These dendrimers are internalized by monocytes within a few seconds, and induce their anti-inflammatory activation. The presence of the dendrimers induces also the inhibition of the differentiation of monocytes into osteoclasts, the maturation of dendritic cells, and inhibits the proliferation of the proinflammatory CD4+ T lymphocytes. Finally, after 2-3 weeks of culture of peripheral blood mononuclear cells, amplifications by several tens of natural killer cells is observed. In view of all these properties, the influence of these azabisphosphonate-dendrimers has been tested in vivo with several animal models, against different chronic or acute inflammatory diseases, such as multiple sclerosis, rheumatoid arthritis, uveitis, and psoriasis, but also against myeloid leukemia, a hematological cancer. The hematological safety has been demonstrated in mice, as there is no platelet aggregation, no hemolysis, and no disturbance in the hematological formula. The safety of the azabisphosphonate-dendrimer has been assessed also with non-human primates (cynomolgus monkeys) which received repeated injections, as a de-risking pre-clinical test. Biochemical, hematological, and all immunological parameters in peripheral blood remained within a normal physiological range throughout the study, and all survived well. Other phosphorous dendrimers also display anti-inflammatory properties in vivo, in particular dendrimers functionalized with mannose derivatives, which prevent acute lung diseases when given orally (per os) to mice. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Anne-Marie Caminade
- Laboratoire de Chimie de Coordination (LCC), CNRS UPR8241, Toulouse Cedex 4, France
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Cédric-Olivier Turrin
- Laboratoire de Chimie de Coordination (LCC), CNRS UPR8241, Toulouse Cedex 4, France
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
- IMD-Pharma, Toulouse Cedex 4, France
| | - Rémy Poupot
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, CHU Purpan, Toulouse Cedex 3, France
- Infinity, Université Toulouse, CNRS, INSERM, UPS, Toulouse, France
| |
Collapse
|
38
|
Roles of Fatty Acids in Microglial Polarization: Evidence from In Vitro and In Vivo Studies on Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23137300. [PMID: 35806302 PMCID: PMC9266841 DOI: 10.3390/ijms23137300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Microglial polarization to the M1 phenotype (classically activated) or the M2 phenotype (alternatively activated) is critical in determining the fate of immune responses in neurodegenerative diseases (NDs). M1 macrophages contribute to neurotoxicity, neuronal and synaptic damage, and oxidative stress and are the first line of defense, and M2 macrophages elicit an anti-inflammatory response to regulate neuroinflammation, clear cell debris, and promote neuroregeneration. Various studies have focused on the ability of natural compounds to promote microglial polarization from the M1 phenotype to the M2 phenotype in several diseases, including NDs. However, studies on the roles of fatty acids in microglial polarization and their implications in NDs are a rare find. Most of the studies support the role of polyunsaturated fatty acids (PUFAs) in microglial polarization using cell and animal models. Thus, we aimed to collect data and provide a narrative account of microglial types, markers, and studies pertaining to fatty acids, particularly PUFAs, on microglial polarization and their neuroprotective effects. The involvement of only PUFAs in the chosen topic necessitates more in-depth research into the role of unexplored fatty acids in microglial polarization and their mechanistic implications. The review also highlights limitations and future challenges.
Collapse
|
39
|
Ghorbani MM, Farazmandfar T, Abediankenari S, Hassannia H, Maleki Z, Shahbazi M. Treatment of EAE mice with Treg, G-MDSC and IL-2: a new insight into cell therapy for multiple sclerosis. Immunotherapy 2022; 14:789-798. [PMID: 35678041 DOI: 10.2217/imt-2021-0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: This study investigates the therapeutic and protective effects of Tregs, myeloid-derived suppressor cells (MDSCs) and IL-2 on multiple sclerosis (MS) disease model. Materials & methods: C57BL/6 mice were immunized to develop an experimental autoimmune encephalomyelitis (EAE) model. We then investigated effects of pre- and post-treatment EAE mice with Tregs, MDSCs and IL-2 on inflammation and demyelination in brain tissue, and on the number of Treg, granulocytic-MDSC and Th-17 cells in spleen. Results: Pre- and post-treatment of EAE mice by Tregs, MDSCs and IL-2 resulted in no weight change, reduced Th-17 cells and suppression of pathological properties. Conclusion: Pre- and post-treatment of immunized mice by Tregs, MDSCs and IL-2 prevent EAE induction.
Collapse
Affiliation(s)
- Mohammad Mehdi Ghorbani
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Touraj Farazmandfar
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Saeid Abediankenari
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hadi Hassannia
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Maleki
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
40
|
Sheu KM, Hoffmann A. Functional Hallmarks of Healthy Macrophage Responses: Their Regulatory Basis and Disease Relevance. Annu Rev Immunol 2022; 40:295-321. [PMID: 35471841 PMCID: PMC10074967 DOI: 10.1146/annurev-immunol-101320-031555] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Macrophages are first responders for the immune system. In this role, they have both effector functions for neutralizing pathogens and sentinel functions for alerting other immune cells of diverse pathologic threats, thereby initiating and coordinating a multipronged immune response. Macrophages are distributed throughout the body-they circulate in the blood, line the mucosal membranes, reside within organs, and survey the connective tissue. Several reviews have summarized their diverse roles in different physiological scenarios and in the initiation or amplification of different pathologies. In this review, we propose that both the effector and the sentinel functions of healthy macrophages rely on three hallmark properties: response specificity, context dependence, and stimulus memory. When these hallmark properties are diminished, the macrophage's biological functions are impaired, which in turn results in increased risk for immune dysregulation, manifested by immune deficiency or autoimmunity. We review the evidence and the molecular mechanisms supporting these functional hallmarks.
Collapse
Affiliation(s)
- Katherine M Sheu
- Department of Microbiology, Immunology, and Molecular Genetics and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, California, USA;
| | - Alexander Hoffmann
- Department of Microbiology, Immunology, and Molecular Genetics and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, California, USA;
| |
Collapse
|
41
|
Frieser D, Pignata A, Khajavi L, Shlesinger D, Gonzalez-Fierro C, Nguyen XH, Yermanos A, Merkler D, Höftberger R, Desestret V, Mair KM, Bauer J, Masson F, Liblau RS. Tissue-resident CD8 + T cells drive compartmentalized and chronic autoimmune damage against CNS neurons. Sci Transl Med 2022; 14:eabl6157. [PMID: 35417189 DOI: 10.1126/scitranslmed.abl6157] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mechanisms underlying the chronicity of autoimmune diseases of the central nervous system (CNS) are largely unknown. In particular, it is unclear whether tissue-resident memory T cells (TRM) contribute to lesion pathogenesis during chronic CNS autoimmunity. Here, we observed that a high frequency of brain-infiltrating CD8+ T cells exhibit a TRM-like phenotype in human autoimmune encephalitis. Using mouse models of neuronal autoimmunity and a combination of T single-cell transcriptomics, high-dimensional flow cytometry, and histopathology, we found that pathogenic CD8+ T cells behind the blood-brain barrier adopt a characteristic TRM differentiation program, and we revealed their phenotypic and functional heterogeneity. In the diseased CNS, autoreactive tissue-resident CD8+ T cells sustained focal neuroinflammation and progressive loss of neurons, independently of recirculating CD8+ T cells. Consistently, a large fraction of autoreactive tissue-resident CD8+ T cells exhibited proliferative potential as well as proinflammatory and cytotoxic properties. Persistence of tissue-resident CD8+ T cells in the CNS and their functional output, but not their initial differentiation, were crucially dependent on CD4+ T cells. Collectively, our results point to tissue-resident CD8+ T cells as essential drivers of chronic CNS autoimmunity and suggest that therapies targeting this compartmentalized autoreactive T cell subset might be effective for treating CNS autoimmune diseases.
Collapse
Affiliation(s)
- David Frieser
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Aurora Pignata
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Leila Khajavi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | | | - Carmen Gonzalez-Fierro
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Xuan-Hung Nguyen
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Alexander Yermanos
- Institute of Microbiology, ETH Zurich, 8093 Zurich, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland.,Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Virginie Desestret
- National Reference Center for Paraneoplastic Neurological Syndromes, MeLiS-UCBL-CNRS, INSERM, Hôpital Neurologique, Hospices Civils de Lyon, 69500 Lyon, France
| | - Katharina M Mair
- Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Jan Bauer
- Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Frederick Masson
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Roland S Liblau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France.,Department of Immunology, Toulouse University Hospital, 31300 Toulouse, France
| |
Collapse
|
42
|
Jiang P, Li X. Regulatory Mechanism of lncRNAs in M1/M2 Macrophages Polarization in the Diseases of Different Etiology. Front Immunol 2022; 13:835932. [PMID: 35145526 PMCID: PMC8822266 DOI: 10.3389/fimmu.2022.835932] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/10/2022] [Indexed: 01/27/2023] Open
Abstract
Precise expression and regulation of genes in the immune system is important for organisms to produce strong immunity towards pathogens and limit autoimmunity. In recent years, an increasing number of studies has shown that long noncoding RNAs (lncRNAs) are closely related to immune function and can participate in regulating immune responses by regulating immune cell differentiation, development, and function. As immune cells, the polarization response of macrophages (Mφs) plays an important role in immune function and inflammation. LncRNAs can regulate the phenotypic polarization of Mφs to M1 or M2 through various mechanisms; promote pro-inflammatory or anti-inflammatory effects; and participate in the pathogenesis of cancers, inflammatory diseases, infections, metabolic diseases, and autoimmune diseases. In addition, it is important to explore the regulatory mechanisms of lncRNAs on the dynamic transition between different Mφs phenotypes. Thus, the regulatory role of lncRNAs in the polarization of Mφs and their mechanism are discussed in this review.
Collapse
Affiliation(s)
- Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaopeng Li
- Department of Neurology, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
- Integrated Traditional Chinese and Western Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Xiaopeng Li,
| |
Collapse
|
43
|
Landi D, Cola G, Mantero V, Balgera R, Moiola L, Nozzolillo A, Dattola V, Sinisi L, Fantozzi R, Di Lemme S, Centonze D, Mataluni G, Nicoletti CG, Marfia GA. Safety of Natalizumab infusion in multiple sclerosis patients during active SARS-CoV-2 infection. Mult Scler Relat Disord 2022; 57:103345. [PMID: 35158454 PMCID: PMC8542399 DOI: 10.1016/j.msard.2021.103345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/16/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022]
Abstract
COVID-19 pandemic represented a challenge in the management of treatments for Multiple Sclerosis (MS), such as Natalizumab (NTZ). NTZ interferes with the homing of lymphocytes into the central nervous system, reducing immune surveillance against opportunistic infection. Although NTZ efficacy starts to decline 8 weeks after the last infusion, increasing the risk of disease reactivation, evidence is lacking on the safety of reinfusion during active SARS-CoV-2 infection. We report clinical outcomes of 18 pwMS receiving NTZ retreatment during confirmed SARS-CoV-2 infection. No worsening of infection or recovery delay was observed. Our data supports the safety of NTZ redosing in these circumstances.
Collapse
Affiliation(s)
- Doriana Landi
- University of Rome Tor Vergata, Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Roma, Italy
| | - Gaia Cola
- University of Rome Tor Vergata, Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Roma, Italy
| | | | | | - Lucia Moiola
- San Raffaele Hospital IRCCS, Multiple sclerosis center-Neurology Department, Milano, Italy
| | - Agostino Nozzolillo
- San Raffaele Hospital IRCCS, Multiple sclerosis center-Neurology Department, Milano, Italy
| | - Vincenzo Dattola
- Grande Ospedale Metropolitano Bianchi Melacrino Morelli, UOC Neurologia, Reggio Calabria, Italy
| | - Leonardo Sinisi
- San Paolo Hospital ASL Napoli 1 Centro, Neurology Unit and MS Center, Napoli, Italy
| | | | | | - Diego Centonze
- IRCCS Neuromed, Unit of Neurology, Pozzilli (IS), Italy; University of Rome Tor Vergata, Department of Systems Medicine, Roma, Italy
| | - Giorgia Mataluni
- University of Rome Tor Vergata, Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Roma, Italy
| | - Carolina Gabri Nicoletti
- University of Rome Tor Vergata, Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Roma, Italy
| | - Girolama Alessandra Marfia
- University of Rome Tor Vergata, Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Roma, Italy; IRCCS Neuromed, Unit of Neurology, Pozzilli (IS), Italy
| |
Collapse
|
44
|
T-Cell Response against Varicella Zoster Virus in Patients with Multiple Sclerosis during Relapse and Remission. Int J Mol Sci 2021; 23:ijms23010298. [PMID: 35008726 PMCID: PMC8745673 DOI: 10.3390/ijms23010298] [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: 09/28/2021] [Revised: 10/31/2021] [Accepted: 11/23/2021] [Indexed: 11/25/2022] Open
Abstract
An association between varicella zoster virus (VZV) and multiple sclerosis (MS) has been reported in Mexican populations. The aim of this study was to compare the response of T cells from MS patients, during relapse and remission, to in vitro stimulation with VZV, adenovirus (AV) and Epstein–Barr virus (EBV). Proliferation and cytokine secretion of T cells from 29 relapsing-remitting MS patients and 38 healthy controls (HC) were analyzed by flow cytometry after stimulating with VZV, AV or EBV. IgG and IgM levels against VZV and EBV were quantified using Enzyme-Linked Immunosorbent Assay. Relapsing MS patients showed a higher percentage of responding CD4+ and CD8+ T cells against VZV compared to AV. In HC and remitting MS patients, proliferation of CD4+ T cells was higher when stimulated with VZV as compared to EBV. Moreover, T cells isolated from remitting patients secreted predominantly Th1 cytokines when cell cultures were stimulated with VZV. Finally, high concentration of anti-VZV IgG was found in sera from patients and controls. The results support previous studies of an VZV-MS association in the particular population studied and provide additional information about the possible role of this virus in the pathogenesis of MS.
Collapse
|
45
|
Role of Phytoconstituents as PPAR Agonists: Implications for Neurodegenerative Disorders. Biomedicines 2021; 9:biomedicines9121914. [PMID: 34944727 PMCID: PMC8698906 DOI: 10.3390/biomedicines9121914] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPAR-γ, PPAR-α, and PPAR-β/δ) are ligand-dependent nuclear receptors that play a critical role in the regulation of hundreds of genes through their activation. Their expression and targeted activation play an important role in the treatment of a variety of diseases, including neurodegenerative, cardiovascular, diabetes, and cancer. In recent years, several reviews have been published describing the therapeutic potential of PPAR agonists (natural or synthetic) in the disorders listed above; however, no comprehensive report defining the role of naturally derived phytoconstituents as PPAR agonists targeting neurodegenerative diseases has been published. This review will focus on the role of phytoconstituents as PPAR agonists and the relevant preclinical studies and mechanistic insights into their neuroprotective effects. Exemplary research includes flavonoids, fatty acids, cannabinoids, curcumin, genistein, capsaicin, and piperine, all of which have been shown to be PPAR agonists either directly or indirectly. Additionally, a few studies have demonstrated the use of clinical samples in in vitro investigations. The role of the fruit fly Drosophila melanogaster as a potential model for studying neurodegenerative diseases has also been highlighted.
Collapse
|
46
|
Gu Y, Zhou H, Yu H, Yang W, Wang B, Qian F, Cheng Y, He S, Zhao X, Zhu L, Zhang Y, Jin M, Lu E. miR-99a regulates CD4 + T cell differentiation and attenuates experimental autoimmune encephalomyelitis by mTOR-mediated glycolysis. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:1173-1185. [PMID: 34820151 PMCID: PMC8598972 DOI: 10.1016/j.omtn.2021.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 07/13/2021] [Indexed: 12/21/2022]
Abstract
Multiple microRNAs exhibit diverse functions to regulate inflammatory and autoimmune diseases. MicroRNA-99a (miR-99a) has been shown to be involved in adipose tissue inflammation and to be downregulated in the inflammatory lesions of autoimmune diseases rheumatoid arthritis and systemic lupus erythematosus. In this study, we found that miR-99a was downregulated in CD4+ T cells from experimental autoimmune encephalomyelitis (EAE) mice, an animal model of multiple sclerosis. Overexpression of miR-99a alleviated EAE development by promoting regulator T cells and inhibiting T helper type 1 (Th1) cell differentiation. Bioinformatics and functional analyses further revealed that the anti-inflammatory effects of miR-99a was attributable to its role in negatively regulating glycolysis reprogramming of CD4+ T cells by targeting the mTOR pathway. Additionally, miR-99a expression was induced by transforming growth factor β (TGF-β) to regulate CD4+ T cell glycolysis and differentiation. Taken together, our results characterize a pivotal role of miR-99a in regulating CD4+ T cell differentiation and glycolysis reprogramming during EAE development, which may indicate that miR-99a is a promising therapeutic target for the amelioration of multiple sclerosis and possibly other autoimmune diseases.
Collapse
Affiliation(s)
- Yuting Gu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.,Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Hong Zhou
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hongshuang Yu
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Wanlin Yang
- Children's Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, Jiangsu 215006, China
| | - Bei Wang
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Fengtao Qian
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yiji Cheng
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shan He
- Children's Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaonan Zhao
- Children's Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, Jiangsu 215006, China
| | - Linqiao Zhu
- Children's Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, Jiangsu 215006, China
| | - Yanyun Zhang
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China.,Children's Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, Jiangsu 215006, China
| | - Min Jin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.,Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| |
Collapse
|
47
|
Murúa SR, Farez MF, Quintana FJ. The Immune Response in Multiple Sclerosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:121-139. [PMID: 34606377 DOI: 10.1146/annurev-pathol-052920-040318] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune, inflammatory, and neurodegenerative disease that affects the central nervous system (CNS). MS is characterized by immune dysregulation, which results in the infiltration of the CNS by immune cells, triggering demyelination, axonal damage, and neurodegeneration. Although the exact causes of MS are not fully understood, genetic and environmental factors are thought to control MS onset and progression. In this article, we review the main immunological mechanisms involved in MS pathogenesis. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sofía Rodríguez Murúa
- Center for Research on Neuroimmunological Diseases (CIEN), Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires 1428, Argentina;
| | - Mauricio F Farez
- Center for Research on Neuroimmunological Diseases (CIEN), Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires 1428, Argentina;
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
48
|
Blomqvist M, Zetterberg H, Blennow K, Månsson JE. Sulfatide in health and disease. The evaluation of sulfatide in cerebrospinal fluid as a possible biomarker for neurodegeneration. Mol Cell Neurosci 2021; 116:103670. [PMID: 34562592 DOI: 10.1016/j.mcn.2021.103670] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 10/20/2022] Open
Abstract
Sulfatide (3-O-sulfogalactosylceramide, SM4) is a glycosphingolipid, highly multifunctional and particularly enriched in the myelin sheath of neurons. The role of sulfatide has been implicated in various biological fields such as the nervous system, immune system, host-pathogen recognition and infection, beta cell function and haemostasis/thrombosis. Thus, alterations in sulfatide metabolism and production are associated with several human diseases such as neurological and immunological disorders and cancers. The unique lipid-rich composition of myelin reflects the importance of lipids in this specific membrane structure. Sulfatide has been shown to be involved in the regulation of oligodendrocyte differentiation and in the maintenance of the myelin sheath by influencing membrane dynamics involving sorting and lateral assembly of myelin proteins as well as ion channels. Sulfatide is furthermore essential for proper formation of the axo-glial junctions at the paranode together with axonal glycosphingolipids. Alterations in sulfatide metabolism are suggested to contribute to myelin deterioration as well as synaptic dysfunction, neurological decline and inflammation observed in different conditions associated with myelin pathology (mouse models and human disorders). Body fluid biomarkers are of importance for clinical diagnostics as well as for patient stratification in clinical trials and treatment monitoring. Cerebrospinal fluid (CSF) is commonly used as an indirect measure of brain metabolism and analysis of CSF sulfatide might provide information regarding whether the lipid disruption observed in neurodegenerative disorders is reflected in this body fluid. In this review, we evaluate the diagnostic utility of CSF sulfatide as a biomarker for neurodegenerative disorders associated with dysmyelination/demyelination by summarising the current literature on this topic. We can conclude that neither CSF sulfatide levels nor individual sulfatide species consistently reflect the lipid disruption observed in many of the demyelinating disorders. One exception is the lysosomal storage disorder metachromatic leukodystrophy, possibly due to the genetically determined accumulation of non-metabolised sulfatide. We also discuss possible explanations as to why myelin pathology in brain tissue is poorly reflected by the CSF sulfatide concentration. The previous suggestion that CSF sulfatide is a marker of myelin damage has thereby been challenged by more recent studies using more sophisticated laboratory techniques for sulfatide analysis as well as improved sample selection criteria due to increased knowledge on disease pathology.
Collapse
Affiliation(s)
- Maria Blomqvist
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jan-Eric Månsson
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
49
|
Murakami K, Tanaka Y, Murakami M. The gateway reflex: breaking through the blood barriers. Int Immunol 2021; 33:743-748. [PMID: 34505147 DOI: 10.1093/intimm/dxab064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
We have been studying inflammatory diseases, with a special focus on IL-6, and discovered two concepts related to inflammation development. One is the gateway reflex, which is induced by the activation of specific neural circuits followed by establishing gateways for autoreactive CD4 + T cells to pass through blood barriers toward the central nervous system (CNS) and retina during tissue-specific inflammatory diseases. We found that the formation of these gateways is dependent on the IL-6 amplifier, which is machinery for enhanced NF-κB activation in endothelial cells at specific sites. We have found five gateway reflexes in total. Here, we introduce the gateway reflex and the IL-6 amplifier.
Collapse
Affiliation(s)
- Kaoru Murakami
- Division of Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Nishi 7, Kita 15 jo, Kita-ku, Sapporo 060-0808, Hokkaido, Japan
| | - Yuki Tanaka
- Division of Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Nishi 7, Kita 15 jo, Kita-ku, Sapporo 060-0808, Hokkaido, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| | - Masaaki Murakami
- Division of Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Nishi 7, Kita 15 jo, Kita-ku, Sapporo 060-0808, Hokkaido, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| |
Collapse
|
50
|
Wei L, Xue Z, Lan B, Yuan S, Li Y, Guo C, Zhang R, Ding R, Shen H. Arctigenin Exerts Neuroprotective Effect by Ameliorating Cortical Activities in Experimental Autoimmune Encephalomyelitis In Vivo. Front Immunol 2021; 12:691590. [PMID: 34349758 PMCID: PMC8327179 DOI: 10.3389/fimmu.2021.691590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/01/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic disease in the central nervous system (CNS), characterized by inflammatory cells that invade into the brain and the spinal cord. Among a bulk of different MS models, the most widely used and best understood rodent model is experimental autoimmune encephalomyelitis (EAE). Arctigenin, a botanical extract from Arctium lappa, is reported to exhibit pharmacological properties, including anti-inflammation and neuroprotection. However, the effects of arctigenin on neural activity attacked by inflammation in MS are still unclear. Here, we use two-photon calcium imaging to observe the activity of somatosensory cortex neurons in awake EAE mice in vivo and found added hyperactive cells, calcium influx, network connectivity, and synchronization, mainly at preclinical stage of EAE model. Besides, more silent cells and decreased calcium influx and reduced network synchronization accompanied by a compensatory rise in functional connectivity are found at the remission stage. Arctigenin treatment not only restricts inordinate individually neural spiking, calcium influx, and network activity at preclinical stage but also restores neuronal activity and communication at remission stage. In addition, we confirm that the frequency of AMPA receptor-mediated spontaneous excitatory postsynaptic current (sEPSC) is also increased at preclinical stage and can be blunted by arctigenin. These findings suggest that excitotoxicity characterized by calcium influx is involved in EAE at preclinical stage. What is more, arctigenin exerts neuroprotective effect by limiting hyperactivity at preclinical stage and ameliorates EAE symptoms, indicating that arctigenin could be a potential therapeutic drug for neuroprotection in MS-related neuropsychological disorders.
Collapse
Affiliation(s)
- Liangpeng Wei
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Zhenyi Xue
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Baihui Lan
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Shiyang Yuan
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Yuanyuan Li
- Innovation Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cunle Guo
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Rongxin Zhang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ran Ding
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Hui Shen
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
- Research Institute of Neurology, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|