1
|
Zeng H, Xu L, Liu J, Mo L, Li M, Song S, Xu X, Miao S, Zhao M, Yang P. Regulation of Tert methylation alleviates food allergy via regulating the Tert-IL10 signal pathway. Immunol Res 2024; 72:1018-1029. [PMID: 39034374 DOI: 10.1007/s12026-024-09504-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/03/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND The cause of food allergy (FA) is still a mystery. Telomerases are involved in the regulation of immune responses. This study aims to gain an understanding of the contribution of telomerase reverse transcriptase (TERT) to the pathogenesis of FA. METHODS A murine FA model was established with ovalbumin as the specific antigen. The role of TERT in regulating dendritic cell (DC) immune tolerogenic functions was evaluated in this murine model. RESULTS We observed that the Tert promoter was at demethylation status and the Tert expression was elevated in DCs of FA mice. The Tert expression in DCs had a positive correlation with the FA response. TERT prevented the induction of Il10 expression in DCs. The immune tolerogenic functions of DCs were diminished by TERT. The immune tolerogenic functions of DC were restored by CpG by boosting the Tert promoter methylation. Administration of CpG promoted the therapeutic effects of allergen specific immunotherapy in FA mice. CONCLUSIONS Low levels of Il10 expression and high levels of Tert expression were observed in intestinal DCs of FA mice. CpG exposure restored the expression of Il10 and increased the therapeutic benefits of allergen-specific immunotherapy.
Collapse
Affiliation(s)
- Haotao Zeng
- Department of Allergy, Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
| | - Lingzhi Xu
- Department of Immunology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Jiangqi Liu
- Department of Allergy, Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
| | - Lihua Mo
- State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen University and Institute of Allergy & Immunology of Shenzhen University, Room A7-509 at Lihu Campus of Shenzhen University. 1066 Xueyuan Blvd, Shenzhen, 518055, China
- Department of General Practice Medicine, Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| | - Minyao Li
- State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen University and Institute of Allergy & Immunology of Shenzhen University, Room A7-509 at Lihu Campus of Shenzhen University. 1066 Xueyuan Blvd, Shenzhen, 518055, China
- Department of General Practice Medicine, Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| | - Shuo Song
- State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen University and Institute of Allergy & Immunology of Shenzhen University, Room A7-509 at Lihu Campus of Shenzhen University. 1066 Xueyuan Blvd, Shenzhen, 518055, China
- Department of General Practice Medicine, Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| | - Xuejie Xu
- State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen University and Institute of Allergy & Immunology of Shenzhen University, Room A7-509 at Lihu Campus of Shenzhen University. 1066 Xueyuan Blvd, Shenzhen, 518055, China
| | - Shihan Miao
- Shenzhen Senior High School Group, Shenzhen, China
| | - Miao Zhao
- Department of Allergy, Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China.
| | - Pingchang Yang
- State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen University and Institute of Allergy & Immunology of Shenzhen University, Room A7-509 at Lihu Campus of Shenzhen University. 1066 Xueyuan Blvd, Shenzhen, 518055, China.
| |
Collapse
|
2
|
Hwang M, Bergmann CC. Neurotropic murine coronavirus mediated demyelination: Factors dampening pathogenesis. J Neuroimmunol 2024; 393:578382. [PMID: 38850674 DOI: 10.1016/j.jneuroim.2024.578382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/12/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Virus infections and autoimmune responses are implicated as primary triggers of demyelinating diseases. Specifically, the association of Epstein-Barr virus (EBV) infection with development of multiple sclerosis (MS) has re-ignited an interest in virus induced autoimmune responses to CNS antigens. Nevertheless, demyelination may also be caused by immune mediated bystander pathology in an attempt to control direct infection in the CNS. Tissue damage as a result of anti-viral responses or low level viral persistence may lead to immune activation manifesting in demyelinating lesions, axonal damage and clinical symptoms. This review focuses on the neurotropic mouse coronavirus induced demyelination model to highlight how immune responses activated during the acute phase pave the way to dampen pathology and promote repair. We specifically discuss the role of immune dampening factors programmed cell death ligand 1 (PD-L1) and interleukin (IL)-10, as well as microglia and triggering receptor expressed on myeloid cells 2 (Trem2), in limiting demyelination independent of viral persistence.
Collapse
Affiliation(s)
- Mihyun Hwang
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Cornelia C Bergmann
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
3
|
Jalali Kondori B, Abdolmaleki A, Raei M, Ghorbani Alvanegh A, Esmaeili Gouvarchin Ghaleh H. Intraperitoneal injection of mesenchymal stem cells-conditioned media (MSCS-CM) treated monocyte can potentially alleviate motor defects in experimental autoimmune encephalomyelitis female mice; An original experimental study. Transpl Immunol 2024; 85:102067. [PMID: 38839021 DOI: 10.1016/j.trim.2024.102067] [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: 09/29/2023] [Revised: 05/25/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
INTRODUCTION Multiple sclerosis (MS), as a destructive pathology of myelin in central nervous system (CNS), causes physical and mental complications. Experimental autoimmune encephalomyelitis (EAE) is laboratory model of MS widely used for CNS-associated inflammatory researches. Cell therapy using macrophage M2 (MPM2) is a cell type with anti-inflammatory characteristics for all inflammatory-based neuropathies. This experimental study investigated the probable therapeutic anti-inflammatory effects of intraperitoneal (IP) injection of MPM2 on alleviation of motor defect in EAE-affected animals. MATERIALS AND METHODS 24 C57/BL6 female mice were divided into four groups of EAE, EAE + Dexa, EAE + PBS, and EAE + MP2. EAE was induced through deep cervical injection of spinal homogenate of guinea pigs. MPM2 cells were harvested from bone marrow and injected (106cells/ml) in three days of 10, 13 and 16 post-immunizations (p.i). Clinical score (CS), anti-inflammatory cytokines (IL-4, IL-10), pro-inflammatory gene expression (TNF-α, IL-1β) and histopathological investigations (HE, Nissl and Luxol Fast Blue) were considered. Data were analyzed using SPSS software (v.19) and p < 0.05 was considered significant level. RESULTS During EAE induction, the mean animal weight was decreased (p < 0.05); besides, following MPM2 injection, the weight gain was applied (p < 0.05) in EAE + MPM2 groups than control. Increased (p < 0.05) levels of CS was found during EAE induction in days 17-28 in EAE animals; besides, CS was decreased (p < 0.05) in EAE + MPM2 group than EAE animals. Also, in days 25-28 of experiment, the CS was decreased (p < 0.05) in EAE + MPM2 than EAE + Dexa. Histopathological assessments revealed low density of cell nuclei in corpus callosum, microscopically. LFB staining also showed considerable decrease in white matter density of corpus callosum in EAE group. Acceleration of white matter density was found in EAE + MPM2 group following cell therapy procedure. Genes expression of TNF-α, IL-1β along with IL-4 and IL-10 were decreased (p < 0.05) in EAE + MPM2 group. CONCLUSION IP injection of MPM2 to EAE-affected female mice can potentially reduce the CNS inflammation, neuronal death and myelin destruction. MPM2 cell therapy can improve animal motor defects.
Collapse
Affiliation(s)
- Bahman Jalali Kondori
- Department of Anatomical Sciences, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran; Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Amir Abdolmaleki
- Department of Anatomical Sciences, Medical School, Baqiyatallah University of Medical Sciences, Tehran, Iran; Department of Operating Room, Nahavand School of Allied Medical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Mahdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | | | - Hadi Esmaeili Gouvarchin Ghaleh
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Aliyu M, Zohora FT, Ceylan A, Hossain F, Yazdani R, Azizi G. Immunopathogenesis of multiple sclerosis: molecular and cellular mechanisms and new immunotherapeutic approaches. Immunopharmacol Immunotoxicol 2024; 46:355-377. [PMID: 38634438 DOI: 10.1080/08923973.2024.2330642] [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: 09/21/2023] [Accepted: 03/09/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a central nervous system (CNS) demyelinating autoimmune disease with increasing global prevalence. It predominantly affects females, especially those of European descent. The interplay between environmental factors and genetic predisposition plays a crucial role in MS etiopathogenesis. METHODS We searched recent relevant literature on reputable databases, which include, PubMed, Embase, Web of Science, Scopus, and ScienceDirect using the following keywords: multiple sclerosis, pathogenesis, autoimmunity, demyelination, therapy, and immunotherapy. RESULTS Various animal models have been employed to investigate the MS etiopathogenesis and therapeutics. Autoreactive T cells within the CNS recruit myeloid cells through chemokine expression, leading to the secretion of inflammatory cytokines driving the MS pathogenesis, resulting in demyelination, gliosis, and axonal loss. Key players include T cell lymphocytes (CD4+ and CD8+), B cells, and neutrophils. Signaling dysregulation in inflammatory pathways and the immunogenetic basis of MS are essential considerations for any successful therapy to MS. Data indicates that B cells and neutrophils also have significant roles in MS, despite the common belief that T cells are essential. High neutrophil-to-lymphocyte ratios correlate with MS severity, indicating their contribution to disease progression. Dysregulated signaling pathways further exacerbate MS progression. CONCLUSION MS remains incurable, but disease-modifying therapies, monoclonal antibodies, and immunomodulatory drugs offer hope for patients. Research on the immunogenetics and immunoregulatory functions of gut microbiota is continuing to provide light on possible treatment avenues. Understanding the complex interplay between genetic predisposition, environmental factors, and immune dysregulation is critical for developing effective treatments for MS.
Collapse
Affiliation(s)
- Mansur Aliyu
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, International Campus, TUMS-IC, Tehran, Iran
- Department of Medical Microbiology, Faculty of Clinical Science, College of Health Sciences, Bayero University, Kano, Nigeria
| | - Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ayca Ceylan
- Medical Faculty, Department of Pediatrics, Division of Immunology and Allergy, Selcuk University, Konya, Turkey
| | - Fariha Hossain
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Reza Yazdani
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gholamreza Azizi
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
5
|
Dolcetti E, Buttari F, Bruno A, Azzolini F, Gilio L, Di Caprio V, Lauritano G, Borrelli A, Galifi G, Furlan R, Finardi A, Musella A, Guadalupi L, Mandolesi G, Rovella V, Centonze D, Stampanoni Bassi M. Low-contrast visual acuity test is associated with central inflammation and predicts disability development in newly diagnosed multiple sclerosis patients. Front Neurol 2024; 15:1326506. [PMID: 38585351 PMCID: PMC10995923 DOI: 10.3389/fneur.2024.1326506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/14/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction The visual system is a prominent site of damage in MS since the earliest phases of the disease. Altered low-contrast visual acuity (LCVA) test has been associated with visual impairment and retinal degeneration, predicting medium- and long-term disability. However, it is unclear whether LCVA may also represent a reliable measure of neuroinflammation and a predictor of disease evolution in the very early stages of MS. Methods We explored in a group of 76 consecutive newly diagnosed relapsing-remitting MS (RR-MS) patients without visual impairment or altered visual evoked potentials, the association between LCVA scores at 2.5% and 1.25% and clinical characteristics, including prospective disability evaluated after 1- and 2 years of follow-up. Associations between LCVA and the CSF levels of IL-10 at diagnosis were also analyzed. Results A negative correlation was found between LCVA at 2.5% and Expanded Disability Status Scale (EDSS) evaluated at first (Spearman's Rho = -0.349, p = 0.005, n = 62) and second year (Spearman's Rho = -0.418, p < 0.001, n = 62) of follow-up, and negative correlations were found with Multiple Sclerosis Severity Score (MSSS) at first (Spearman's Rho = -0.359, p = 0.004, n = 62) and second year (Spearman's Rho = -0.472, p < 0.001, n = 62). All the data were confirmed by a mixed effect model, considering other clinical variables. A positive correlation was found between the CSF concentrations of IL-10 and LCVA at 2.5% (Spearman's Rho = 0.272, p = 0.020, n = 76), and 1.25% (Spearman's Rho, = 0.276, p = 0.018, n = 76), also evidenced in a linear regression. Discussion In MS patients at diagnosis, altered LCVA may be associated with CSF inflammation and represent a useful parameter to identify patients with worse disease course.
Collapse
Affiliation(s)
| | - Fabio Buttari
- Neurology Unit, IRCSS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | | | | - Luana Gilio
- Neurology Unit, IRCSS Neuromed, Pozzilli, Italy
- Faculty of Psychology, Uninettuno Telematic International University, Rome, Italy
| | | | | | | | | | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| | - Livia Guadalupi
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| | | | - Diego Centonze
- Neurology Unit, IRCSS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | |
Collapse
|
6
|
Holcomb L, Holman JM, Hurd M, Lavoie B, Colucci L, Hunt B, Hunt T, Kinney M, Pathak J, Mawe GM, Moses PL, Perry E, Stratigakis A, Zhang T, Chen G, Ishaq SL, Li Y. Early life exposure to broccoli sprouts confers stronger protection against enterocolitis development in an immunological mouse model of inflammatory bowel disease. mSystems 2023; 8:e0068823. [PMID: 37942948 PMCID: PMC10734470 DOI: 10.1128/msystems.00688-23] [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: 07/21/2023] [Accepted: 10/02/2023] [Indexed: 11/10/2023] Open
Abstract
IMPORTANCE To our knowledge, IL-10-KO mice have not previously been used to investigate the interactions of host, microbiota, and broccoli, broccoli sprouts, or broccoli bioactives in resolving symptoms of CD. We showed that a diet containing 10% raw broccoli sprouts increased the plasma concentration of the anti-inflammatory compound sulforaphane and protected mice to varying degrees against disease symptoms, including weight loss or stagnation, fecal blood, and diarrhea. Younger mice responded more strongly to the diet, further reducing symptoms, as well as increased gut bacterial richness, increased bacterial community similarity to each other, and more location-specific communities than older mice on the diet intervention. Crohn's disease disrupts the lives of patients and requires people to alter dietary and lifestyle habits to manage symptoms. The current medical treatment is expensive with significant side effects, and a dietary intervention represents an affordable, accessible, and simple strategy to reduce the burden of symptoms.
Collapse
Affiliation(s)
- Lola Holcomb
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| | - Johanna M. Holman
- School of Food and Agriculture, University of Maine, Orono, Maine, USA
| | - Molly Hurd
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Brigitte Lavoie
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Louisa Colucci
- Department of Biology, Husson University, Bangor, Maine, USA
| | - Benjamin Hunt
- Department of Biology, University of Maine, Orono, Maine, USA
| | - Timothy Hunt
- Department of Biology, University of Maine, Orono, Maine, USA
| | - Marissa Kinney
- School of Food and Agriculture, University of Maine, Orono, Maine, USA
| | - Jahnavi Pathak
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| | - Gary M. Mawe
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Peter L. Moses
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
- Finch Therapeutics, Somerville, Massachusetts, USA
| | - Emma Perry
- Electron Microscopy Laboratory, University of Maine, Orono, Maine, USA
| | - Allesandra Stratigakis
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Johnson City, New York, USA
| | - Tao Zhang
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Johnson City, New York, USA
| | - Grace Chen
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Suzanne L. Ishaq
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| | - Yanyan Li
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| |
Collapse
|
7
|
Daei Sorkhabi A, Komijani E, Sarkesh A, Ghaderi Shadbad P, Aghebati-Maleki A, Aghebati-Maleki L. Advances in immune checkpoint-based immunotherapies for multiple sclerosis: rationale and practice. Cell Commun Signal 2023; 21:321. [PMID: 37946301 PMCID: PMC10634124 DOI: 10.1186/s12964-023-01289-9] [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: 05/11/2023] [Accepted: 08/19/2023] [Indexed: 11/12/2023] Open
Abstract
Beyond the encouraging results and broad clinical applicability of immune checkpoint (ICP) inhibitors in cancer therapy, ICP-based immunotherapies in the context of autoimmune disease, particularly multiple sclerosis (MS), have garnered considerable attention and hold great potential for developing effective therapeutic strategies. Given the well-established immunoregulatory role of ICPs in maintaining a balance between stimulatory and inhibitory signaling pathways to promote immune tolerance to self-antigens, a dysregulated expression pattern of ICPs has been observed in a significant proportion of patients with MS and its animal model called experimental autoimmune encephalomyelitis (EAE), which is associated with autoreactivity towards myelin and neurodegeneration. Consequently, there is a rationale for developing immunotherapeutic strategies to induce inhibitory ICPs while suppressing stimulatory ICPs, including engineering immune cells to overexpress ligands for inhibitory ICP receptors, such as program death-1 (PD-1), or designing fusion proteins, namely abatacept, to bind and inhibit the co-stimulatory pathways involved in overactivated T-cell mediated autoimmunity, and other strategies that will be discussed in-depth in the current review. Video Abstract.
Collapse
Affiliation(s)
- Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Erfan Komijani
- Department of Veterinary, Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pedram Ghaderi Shadbad
- Department of Veterinary, Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Holcomb L, Holman JM, Hurd M, Lavoie B, Colucci L, Hunt B, Hunt T, Kinney M, Pathak J, Mawe GM, Moses PL, Perry E, Stratigakis A, Zhang T, Chen G, Ishaq SL, Li Y. Early life exposure to broccoli sprouts confers stronger protection against enterocolitis development in an immunological mouse model of inflammatory bowel disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.27.525953. [PMID: 36747766 PMCID: PMC9900910 DOI: 10.1101/2023.01.27.525953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Crohn's Disease (CD) is a presentation of Inflammatory Bowel Disease (IBD) that manifests in childhood and adolescence, and involves chronic and severe enterocolitis, immune and gut microbiome dysregulation, and other complications. Diet and gut-microbiota-produced metabolites are sources of anti-inflammatories which could ameliorate symptoms. However, questions remain on how IBD influences biogeographic patterns of microbial location and function in the gut, how early life transitional gut communities are affected by IBD and diet interventions, and how disruption to biogeography alters disease mediation by diet components or microbial metabolites. Many studies on diet and IBD use a chemically induced ulcerative colitis model, despite the availability of an immune-modulated CD model. Interleukin-10-knockout (IL-10-KO) mice on a C57BL/6 background, beginning at age 4 or 7 weeks, were fed a control diet or one containing 10% (w/w) raw broccoli sprouts, which was high in the sprout-sourced anti-inflammatory sulforaphane. Diets began 7 days prior to, and for 2 weeks after inoculation with Helicobacter hepaticus, which triggers Crohn's-like symptoms in these immune-impaired mice. The broccoli sprout diet increased sulforaphane in plasma; decreased weight stagnation, fecal blood, and diarrhea associated; and increased microbiota richness in the gut, especially in younger mice. Sprout diets resulted in some anatomically specific bacteria in younger mice, and reduced the prevalence and abundance of pathobiont bacteria which trigger inflammation in the IL-10-KO mouse, for example; Escherichia coli and Helicobacter. Overall, the IL-10-KO mouse model is responsive to a raw broccoli sprout diet and represents an opportunity for more diet-host-microbiome research.
Collapse
Affiliation(s)
- Lola Holcomb
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA 04469
| | - Johanna M. Holman
- School of Food and Agriculture, University of Maine, Orono, Maine, USA 04469
| | - Molly Hurd
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA 05401
| | - Brigitte Lavoie
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA 05401
| | - Louisa Colucci
- Department of Biology, Husson University, Bangor, Maine, USA 04401
| | - Benjamin Hunt
- Department of Biology, University of Maine, Orono, Maine, USA 04469
| | - Timothy Hunt
- Department of Biology, University of Maine, Orono, Maine, USA 04469
| | - Marissa Kinney
- School of Food and Agriculture, University of Maine, Orono, Maine, USA 04469
| | - Jahnavi Pathak
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA 04469
| | - Gary M. Mawe
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA 05401
| | - Peter L. Moses
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA 05401
- Finch Therapeutics, Somerville, Massachusetts, USA 02143
| | - Emma Perry
- Electron Microscopy Laboratory, University of Maine, Orono, Maine, USA 04469
| | - Allesandra Stratigakis
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Johnson City, New York, USA 13790
| | - Tao Zhang
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Johnson City, New York, USA 13790
| | - Grace Chen
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA 48109
| | - Suzanne L. Ishaq
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA 04469
| | - Yanyan Li
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA 04469
| |
Collapse
|
9
|
Offner H, Lockwood D, Meza-Romero R, Vandenbark AA. PD-L1 is required for estrogen-induced protection against severe EAE in IL-10 deficient mice 1. Metab Brain Dis 2023; 38:589-599. [PMID: 36454506 PMCID: PMC9976593 DOI: 10.1007/s11011-022-01129-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND IL-10 knockout (KO) mice can be protected against experimental autoimmune encephalomyelitis (EAE) with low-dose estrogen (E2) treatment similar to wild type (WT) mice, indicating that IL-10 is not required for E2-induced EAE protection. Our previous study demonstrated that E2 treatment induced an increase in programmed death ligands 1 (PD-L1) and 2 (PD-L2) on monocytes and macrophages in the periphery and within the CNS. In this study, we selectively inhibited the function of PD-L1 and PD-L2 to evaluate their critical role in maintaining E2-induced protection against EAE in IL-10-KO mice. METHODS This study used female IL-10 KO mice pre-treated with either E2 or sham pellets seven days prior to induction of EAE and subsequently treated with Vehicle or antibodies to PD-L1, PD-L2 or respective isotype controls. Mice were scored daily for EAE severity over 21 days post-EAE induction. Cells from the spleen and brain were evaluated by flow cytometry. RESULTS Differences in EAE severity were assessed in E2 and sham pre-treated IL-10-KO mice treated with α-PD-L1 or α-PD-L2 antibodies over the course of disease compared to treatment with Vehicle or isotype control antibodies. The results revealed real-time development of severe EAE in E2-pre-treated IL-10-KO mice treated with α-PD-L1 but not α-PD-L2 antibodies, mediated in part by increased percentages of activated CD74+CD11b+ myeloid cells in spleen and brain as well as splenic B-cells, T-cells and CD73+ cells. CONCLUSION These results demonstrate unequivocally that PD-L1 but not PD-L2 was required to retain the inhibitory effects of E2 on clinical EAE scores in female IL-10-KO mice and further implicate the emergence of the MIF/CD74 axis as a contributing pathogenic mechanism.
Collapse
Affiliation(s)
- Halina Offner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA.
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.
- Veterans Affairs Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., 97239, Portland, OR, USA.
| | - Denesa Lockwood
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
| | - Roberto Meza-Romero
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
10
|
Hasan Z, Nguyen TQ, Lam BWS, Wong JHX, Wong CCY, Tan CKH, Yu J, Thiam CH, Zhang Y, Angeli V, Nguyen LN. Postnatal deletion of Spns2 prevents neuroinflammation without compromising blood vascular functions. Cell Mol Life Sci 2022; 79:541. [PMID: 36198832 DOI: 10.1007/s00018-022-04573-y] [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: 01/04/2022] [Revised: 09/09/2022] [Accepted: 09/27/2022] [Indexed: 11/03/2022]
Abstract
Protein Spinster homolog 2 (Spns2) is a sphingosine-1-phosphate (S1P) transporter that releases S1P to regulate lymphocyte egress and trafficking. Global deletion of Spns2 (Spns2-/-) has been shown to reduce disease severity in several autoimmune disease models. To examine whether Spns2 could be exploited as a drug target, we generated and characterized the mice with postnatal knockout of Spns2 (Spns2-Mx1Cre). Our results showed that Spns2-Mx1Cre mice had significantly low number of lymphocytes in blood and lymphoid organs similar to Spns2-/- mice. Lymph but not plasma S1P levels were significantly reduced in both groups of knockout mice. Our lipidomic results also showed that Spns2 releases different S1P species into lymph. Interestingly, lymphatic vessels in the lymph nodes (LNs) of Spns2-/- and Spns2-Mx1Cre mice exhibited morphological defects. The structures of high endothelial venules (HEV) in the LNs of Spns2-Mx1Cre mice were disorganized. These results indicate that lack of Spns2 affects both S1P secretion and LN vasculatures. Nevertheless, blood vasculature of these Spns2 deficient mice was not different to controls under homeostasis and vascular insults. Importantly, Spns2-Mx1Cre mice were resistant to multiple sclerosis in experimental autoimmune encephalomyelitis (EAE) models with significant reduction of pathogenic Th17 cells in the central nervous system (CNS). This study suggests that pharmacological inhibition of Spns2 may be exploited for therapeutic applications in treatment of neuroinflammation.
Collapse
Affiliation(s)
- Zafrul Hasan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Toan Q Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Brenda Wan Shing Lam
- Department of Pharmacology, Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, Singapore
| | - Jovi Hui Xin Wong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Caleb Cheng Yi Wong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Clarissa Kai Hui Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Jiabo Yu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore.,Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Chung Hwee Thiam
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore.,Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore.,Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Veronique Angeli
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore.,Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore. .,Life Sciences Institute, Singapore Lipidomics Incubator (SLING), National University of Singapore, Singapore, 117456, Singapore. .,Cardiovascular Disease Research (CVD) Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore. .,Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore. .,Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
11
|
Edo Á, Calvo-Barreiro L, Eixarch H, Bosch A, Chillón M, Espejo C. Therapeutic Effect of IL-21 Blockage by Gene Therapy in Experimental Autoimmune Encephalomyelitis. Neurotherapeutics 2022; 19:1617-1633. [PMID: 35902536 PMCID: PMC9606180 DOI: 10.1007/s13311-022-01279-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
The pathogenic role of the interleukin 21 (IL-21) in different autoimmune diseases, such as multiple sclerosis (MS), has been extensively studied. However, its pleiotropic nature makes it a cytokine that may exhibit different activity depending on the immunological stage of the disease. In this study, we developed a gene therapy strategy to block the interaction between IL-21 and its receptor (IL-21R) by using adeno-associated vectors (AAV) encoding a new soluble cytokine receptor (sIL21R) protein. We tested this strategy in a murine model of experimental autoimmune encephalomyelitis (EAE), obtaining different clinical effects depending on the time at which the treatment was applied. Although the administration of the treatment during the development of the immune response was counterproductive, the preventive administration of the therapeutic vectors showed a protective effect by reducing the number of animals that developed the disease, as well as an improvement at the histopathological level and a modification of the immunological profile of the animals treated with the AAV8.sIL21R. The beneficial effect of the treatment was also observed when inducing the expression of the therapeutic molecule once the first neurological signs were established in a therapeutic approach with a doxycyline (Dox)-inducible expression system. All these clinical results highlight the pleiotropicity of this cytokine in the different clinical stages and its key role in the EAE immunopathogenesis.
Collapse
Affiliation(s)
- Ángel Edo
- Institut de Neurociències (INc), Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, (Campus UAB), Bellaterra, Cerdanyola del Vallès (Ed. H 5th level), 08193, Barcelona, Spain
- Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Laura Calvo-Barreiro
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Pg. Vall d'Hebron 119-129 (Ed. Collserola, Lab. 149), 08035, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Herena Eixarch
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Pg. Vall d'Hebron 119-129 (Ed. Collserola, Lab. 149), 08035, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Assumpció Bosch
- Institut de Neurociències (INc), Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, (Campus UAB), Bellaterra, Cerdanyola del Vallès (Ed. H 5th level), 08193, Barcelona, Spain
- Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Miguel Chillón
- Institut de Neurociències (INc), Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, (Campus UAB), Bellaterra, Cerdanyola del Vallès (Ed. H 5th level), 08193, Barcelona, Spain.
- Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
- Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain.
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain.
- Vector Production Unit (UPV), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Carmen Espejo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Pg. Vall d'Hebron 119-129 (Ed. Collserola, Lab. 149), 08035, Barcelona, Spain.
- Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain.
| |
Collapse
|
12
|
Ling X, Wang T, Han C, Wang P, Liu X, Zheng C, Bi J, Zhou X. IFN-γ-Primed hUCMSCs Significantly Reduced Inflammation via the Foxp3/ROR-γt/STAT3 Signaling Pathway in an Animal Model of Multiple Sclerosis. Front Immunol 2022; 13:835345. [PMID: 35300342 PMCID: PMC8921983 DOI: 10.3389/fimmu.2022.835345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Our previous study showed that interferon gamma (IFN-γ) might enhance the immunosuppressive properties of mesenchymal stem cells (MSCs) by upregulating the expression of indoleamine 2,3-dioxygenease. Therefore, we treated experimental autoimmune encephalomyelitis (EAE) mice, an animal model of multiple sclerosis (MS), with IFN-γ-primed human umbilical cord MSCs (IFN-γ-hUCMSCs). This study aimed to investigate the potential therapeutic effects of IFN-γ-hUCMSCs transplantation and to identify the biological pathways involved in EAE mice. Firstly, the body weights and clinical scores of EAE mice were recorded before and after treatment. Then, the inflammatory cytokine levels in splenic cell supernatants were quantified by enzyme-linked immunosorbent assay. Finally, the mRNA expression levels of signal transducer and activator of transduction 3 (STAT3), retinoic acid-related orphan receptor gamma t (ROR-γt), and forkhead box P3 (Foxp3) were detected by quantitative reverse transcription polymerase chain reaction. We observed that IFN-γ-hUCMSCs transplantation significantly alleviated body weight loss and decreased the clinical scores of mice. Additionally, IFN-γ-hUCMSCs transplantation could regulate the production of inflammatory cytokines, interleukin (IL)-10 and IL-17, thereby showing more potent treatment efficacy than human umbilical cord MSCs (hUCMSCs) transplantation (p < 0.05). Compared with the EAE group, the expressions of STAT3 and ROR-γt in the transplantation groups were significantly decreased, but the expression of Foxp3 was significantly upregulated in the IFN-γ-hUCMSCs transplantation group compared to that in the hUCMSCs transplantation group. We assumed that IFN-γ-hUCMSCs may affect the balance of T helper 17 (Th17) cells/regulatory T cells (Tregs) through the Foxp3/ROR-γt/STAT3 signaling pathway to reduce the inflammatory response, thereby improving the clinical symptoms of EAE mice. Our study demonstrated that transplantation of IFN-γ-hUCMSCs could reduce inflammation in EAE mice via the Foxp3/ROR-γt/STAT3 signaling pathway, highlighting the therapeutic effects of IFN-γ-hUCMSCs in patients with MS.
Collapse
Affiliation(s)
- Xiao Ling
- Department of Gynaecology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Teng Wang
- Department of Digestive Internal Medicine, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chao Han
- Department of Neurosurgery, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Pin Wang
- Department of Neurology Medicine, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoli Liu
- Department of Hematology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Institute of Biotherapy for Hematological Malignancies, Shandong University, Jinan, China.,Shandong University-Karolinska Institute Collaboration Laboratory for Stem Cell Research, Shandong University, Jinan, China
| | - Chengyun Zheng
- Department of Hematology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Institute of Biotherapy for Hematological Malignancies, Shandong University, Jinan, China.,Shandong University-Karolinska Institute Collaboration Laboratory for Stem Cell Research, Shandong University, Jinan, China
| | - Jianzhong Bi
- Department of Neurology Medicine, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyan Zhou
- Department of Neurology Medicine, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
13
|
Mi Y, Han J, Zhu J, Jin T. Role of the PD-1/PD-L1 Signaling in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis: Recent Insights and Future Directions. Mol Neurobiol 2021; 58:6249-6271. [PMID: 34480337 PMCID: PMC8639577 DOI: 10.1007/s12035-021-02495-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmunity-related chronic demyelination disease of the central nervous system (CNS), causing young disability. Currently, highly specific immunotherapies for MS are still lacking. Programmed cell death 1 (PD-1) is an immunosuppressive co-stimulatory molecule, which is expressed on activated T lymphocytes, B lymphocytes, natural killer cells, and other immune cells. PD-L1, the ligand of PD-1, is expressed on T lymphocytes, B lymphocytes, dendritic cells, and macrophages. PD-1/PD-L1 delivers negative regulatory signals to immune cells, maintaining immune tolerance and inhibiting autoimmunity. This review comprehensively summarizes current insights into the role of PD-1/PD-L1 signaling in MS and its animal model experimental autoimmune encephalomyelitis (EAE). The potentiality of PD-1/PD-L1 as biomarkers or therapeutic targets for MS will also be discussed.
Collapse
Affiliation(s)
- Yan Mi
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Present Address: Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| |
Collapse
|
14
|
Yeung SSH, Ho YS, Chang RCC. The role of meningeal populations of type II innate lymphoid cells in modulating neuroinflammation in neurodegenerative diseases. Exp Mol Med 2021; 53:1251-1267. [PMID: 34489558 PMCID: PMC8492689 DOI: 10.1038/s12276-021-00660-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023] Open
Abstract
Recent research into meningeal lymphatics has revealed a never-before appreciated role of type II innate lymphoid cells (ILC2s) in modulating neuroinflammation in the central nervous system (CNS). To date, the role of ILC2-mediated inflammation in the periphery has been well studied. However, the exact distribution of ILC2s in the CNS and therefore their putative role in modulating neuroinflammation in neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and major depressive disorder (MDD) remain highly elusive. Here, we review the current evidence of ILC2-mediated modulation of neuroinflammatory cues (i.e., IL-33, IL-25, IL-5, IL-13, IL-10, TNFα, and CXCL16-CXCR6) within the CNS, highlight the distribution of ILC2s in both the periphery and CNS, and discuss some challenges associated with cell type-specific targeting that are important for therapeutics. A comprehensive understanding of the roles of ILC2s in mediating and responding to inflammatory cues may provide valuable insight into potential therapeutic strategies for many dementia-related disorders.
Collapse
Affiliation(s)
- Sherry Sin-Hang Yeung
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Yuen-Shan Ho
- grid.16890.360000 0004 1764 6123School of Nursing, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR China
| | - Raymond Chuen-Chung Chang
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,grid.194645.b0000000121742757State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| |
Collapse
|
15
|
Li H, Zheng C, Han J, Zhu J, Liu S, Jin T. PD-1/PD-L1 Axis as a Potential Therapeutic Target for Multiple Sclerosis: A T Cell Perspective. Front Cell Neurosci 2021; 15:716747. [PMID: 34381337 PMCID: PMC8350166 DOI: 10.3389/fncel.2021.716747] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 06/22/2021] [Indexed: 12/19/2022] Open
Abstract
The programmed cell death protein-1/programmed death ligand-1 (PD-1/PD-L1) axis is a widely studied immune checkpoint that modulates signaling pathways related to T cell activation. The use of PD-1/PD-L1 inhibitors is a promising immune therapy strategy for cancer patients. However, individuals treated with PD-1/PD-L1 inhibitors may develop immune-related adverse events due to excessive immune reactions. Multiple sclerosis (MS) is a chronic demyelinating and neurodegenerative disease of the central nervous system. T cells and the PD-1/PD-L1 axis play vital roles in the pathogenesis of MS. A better understanding of the complex relationship between the PD-1/PD-L1 axis and T cells may extend our knowledge of the molecular mechanisms and therapeutic approaches for MS. In this review, we summarize the most recent findings regarding the role of the PD-1/PD-L1 axis in MS and discuss the potential therapeutic strategies to modulate the expression of PD-1/PD-L1 in MS.
Collapse
Affiliation(s)
- HaiXia Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Chao Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jinming Han
- Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Shan Liu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Ibañez-Vega J, Vilchez C, Jimenez K, Guevara C, Burgos PI, Naves R. Cellular and molecular regulation of the programmed death-1/programmed death ligand system and its role in multiple sclerosis and other autoimmune diseases. J Autoimmun 2021; 123:102702. [PMID: 34311143 DOI: 10.1016/j.jaut.2021.102702] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/07/2021] [Accepted: 07/10/2021] [Indexed: 01/12/2023]
Abstract
Programmed Cell Death 1 (PD-1) receptor and its ligands (PD-Ls) are essential to maintain peripheral immune tolerance and to avoid tissue damage. Consequently, altered gene or protein expression of this system of co-inhibitory molecules has been involved in the development of cancer and autoimmunity. Substantial progress has been achieved in the study of the PD-1/PD-Ls system in terms of regulatory mechanisms and therapy. However, the role of the PD-1/PD-Ls pathway in neuroinflammation has been less explored despite being a potential target of treatment for neurodegenerative diseases. Multiple Sclerosis (MS) is the most prevalent, chronic, inflammatory, and autoimmune disease of the central nervous system that leads to demyelination and axonal damage in young adults. Recent studies have highlighted the key role of the PD-1/PD-Ls pathway in inducing a neuroprotective response and restraining T cell activation and neurodegeneration in MS. In this review, we outline the molecular and cellular mechanisms regulating gene expression, protein synthesis and traffic of PD-1/PD-Ls as well as relevant processes that control PD-1/PD-Ls engagement in the immunological synapse between antigen-presenting cells and T cells. Also, we highlight the most recent findings regarding the role of the PD-1/PD-Ls pathway in MS and its murine model, experimental autoimmune encephalomyelitis (EAE), including the contribution of PD-1 expressing follicular helper T (TFH) cells in the pathogenesis of these diseases. In addition, we compare and contrast results found in MS and EAE with evidence reported in other autoimmune diseases and their experimental models, and review PD-1/PD-Ls-targeting therapeutic approaches.
Collapse
Affiliation(s)
- Jorge Ibañez-Vega
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Constanza Vilchez
- Faculty of Natural Sciences, Mathematics and Environment, Universidad Tecnológica Metropolitana, Santiago, Chile
| | - Karin Jimenez
- Faculty of Natural Sciences, Mathematics and Environment, Universidad Tecnológica Metropolitana, Santiago, Chile
| | - Carlos Guevara
- Department of Neurology and Neurosurgery, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Paula I Burgos
- Department of Clinical Immunology and Rheumatology, School of Medicine, Pontificia Universidad Católica de Chile, Chile.
| | - Rodrigo Naves
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
17
|
Zhou X, Liu X, Liu L, Han C, Xie Z, Liu X, Xu Y, Li F, Bi J, Zheng C. Transplantation of IFN-γ Primed hUCMSCs Significantly Improved Outcomes of Experimental Autoimmune Encephalomyelitis in a Mouse Model. Neurochem Res 2020; 45:1510-1517. [PMID: 32172400 DOI: 10.1007/s11064-020-03009-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/18/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022]
Abstract
The aim of this study was to investigate potential therapeutic effects of IFN-γ primed human umbilical cord mesenchymal stem cell (IFN-γ-hUCMSCs) transplantation on experimental autoimmune encephalomyelitis (EAE) in mice. In this study, EAE mouse model was established by MOG35-55 immunization method. Outcomes of the EAE mice in terms of body weight and clinical symptoms were analyzed. Electromyography (EMG) was performed to evaluate nerve conduction. ELISA was applied to quantify inflammatory cytokine levels in serum. Our results showed that IFN-γ could up-regulate protein expression of indoleamine 2, 3-dioxygenease 1 (IDO1), an important molecule released by MSCs to exert their immune suppressive activity (p < 0.01). In this study treatment efficacy for EAE was compared between transplantation of hUCMSCs alone and the IFN-γ-hUCMSCs which were cultured in the presence of IFN-γ for 48 h prior to be harvested for transplantation. Compared with hUCMSCs alone and control (PBS transfusion) group, transplantation of the IFN-γ-hUCMSCs could significantly alleviate the body weight loss and clinical symptoms of EAE mice (p < 0.05). Consistently EMG latency was significantly improved in treatment groups (p < 0.001), and the IFN-γ-hUCMSCs group was even better than the hUCMSCs group (p < 0.05). Moreover, the concentrations of IL-17A and TNF-α in serum of the mice treated by IFN-γ-hUCMSCs were significantly lower than hUCMSCs alone and controls, respectively (p < 0.05). In few of the roles of IL-17A and TNF-α in the pathogenesis of EAE, IFN-γ-hUCMSCs treatment associated-suppression of IL-17A and TNF-α expression may contribute in part to their therapeutic effects on EAE. In sum, our study highlights a great clinical potential of IFN-γ-hUCMSCs for multiple sclerosis (MS) treatment.
Collapse
Affiliation(s)
- Xiaoyan Zhou
- Department of Neurology Medicine, The Second Hospital of Shandong University, 247th of Beiyuan Rd., Jinan, Shandong, China
| | - Xiaoli Liu
- Department of Hematology, Institute of Biotherapy for Hematological Malignancies, The Second Hospital of Shandong University, 247th of Beiyuan Rd., Jinan, Shandong, China.,Shandong University-Karolinska Institute Collaboration Laboratory for Stem Cell Research, Jinan, Shandong, China
| | - Li Liu
- Department of Neurology Medicine, The Second Hospital of Shandong University, 247th of Beiyuan Rd., Jinan, Shandong, China
| | - Chao Han
- Department of Neurosurgery, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Zhaohong Xie
- Department of Neurology Medicine, The Second Hospital of Shandong University, 247th of Beiyuan Rd., Jinan, Shandong, China
| | - Xiangtian Liu
- Department of Neurology Medicine, The Second Hospital of Shandong University, 247th of Beiyuan Rd., Jinan, Shandong, China
| | - Yingying Xu
- Department of Neurology Medicine, The Second Hospital of Shandong University, 247th of Beiyuan Rd., Jinan, Shandong, China
| | - Fan Li
- Department of Neurology Medicine, The Second Hospital of Shandong University, 247th of Beiyuan Rd., Jinan, Shandong, China
| | - Jianzhong Bi
- Department of Neurology Medicine, The Second Hospital of Shandong University, 247th of Beiyuan Rd., Jinan, Shandong, China.
| | - Chengyun Zheng
- Department of Hematology, Institute of Biotherapy for Hematological Malignancies, The Second Hospital of Shandong University, 247th of Beiyuan Rd., Jinan, Shandong, China. .,Shandong University-Karolinska Institute Collaboration Laboratory for Stem Cell Research, Jinan, Shandong, China.
| |
Collapse
|