1
|
Neili NE, AbdelKafi-Koubaa Z, Jebali J, Kaidi K, Sahraoui G, Ahmed MB, Srairi-Abid N, Marrakchi N, Doghri R, ELBini I. Modulation of αv integrins by lebecetin, a viper venom-derived molecule, in experimental neuroinflammation and demyelination models. Sci Rep 2024; 14:22398. [PMID: 39333683 PMCID: PMC11436777 DOI: 10.1038/s41598-024-73259-1] [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: 05/06/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
Several neurodegenerative diseases, such as multiple sclerosis and Parkinson's disease, are linked to alterations in myelin content or structure. Transmembrane receptors such as integrins could be involved in these alterations. In the present study, we investigated the role of αv-integrins in experimental models of neuroinflammation and demyelination with the use of lebecetin (LCT), a C-lectin protein purified from Macrovipera lebetina viper venom, as an αv-integrin modulator. In a model of neuroinflammation, LCT inhibited the upregulation of αv, β3, β5, α5, and β1 integrins, as well as the associated release of pro-inflammatory factor IL-6 and chemokine CXCL-10, and decreased the expression of phosphorylated NfκB. The subsequent "indirect culture" between reactive astrocytes and oligodendrocytes showed a down-regulation of αv and β3 integrins versus upregulation of β1 one, accompanied by a reduced expression of myelin basic protein (MBP). Treatment of oligodendrocytes with LCT rectified the changes in integrin and MBP expression. Through Western blot quantification, LCT was shown to upregulate the expression levels of PI3K and p-mTOR while downregulating expression levels of p-AKT in oligodendrocytes, suggesting the neuroprotective and pro-myelinating effects of LCT may be related to the PI3K/mTor/AKT pathway. Concomitantly, we found that LCT promoted remyelination by tracking the increased expression of MBP in the brains of cuprizone-intoxicated mice. These results point to an involvement of integrins in not only neuroinflammation but demyelination as well. Thus, targeting αv integrins could offer potential therapeutic avenues for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Nour-Elhouda Neili
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Zaineb AbdelKafi-Koubaa
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Saleh Azaiez Institute, Tunis, Tunisia
| | - Jed Jebali
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Khouloud Kaidi
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Ghada Sahraoui
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Saleh Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Melika Ben Ahmed
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections (LR16IPT02), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Najet Srairi-Abid
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Naziha Marrakchi
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Raoudha Doghri
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Saleh Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Ines ELBini
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia.
| |
Collapse
|
2
|
Zamali I, Elbini I, Rekik R, Neili NE, Ben Hamouda W, Ben Hmid A, Doghri R, Ben Ahmed M. Advancing understanding of the role of IL-22 in myelination: insights from the Cuprizone mouse model. Front Neurol 2024; 15:1411143. [PMID: 39040539 PMCID: PMC11260746 DOI: 10.3389/fneur.2024.1411143] [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: 04/02/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Despite significant advancements in the field, the pathophysiology of multiple sclerosis (MS) remains partially understood, with limited therapeutic options available for this debilitating condition. The precise impact of Interleukin-22 (IL-22) in the context of MS is still incompletely elucidated with some evidence suggesting its protective role. To provide a more comprehensive understanding of the role of IL-22, we investigated its effect on remyelination in a mouse model of demyelination induced by Cuprizone. Mice underwent a 6 week regimen of Cuprizone or vehicle, followed or not by intraperitoneal administration of IL-22. Behavioral assessments including tail suspension and inverted screen tests were conducted, alongside histological, histochemical, and quantitative PCR analyses. In Cuprizone-treated mice, IL-22 significantly improved motor and behavioral performance and robustly promoted remyelination in the corpus callosum. Additionally, IL-22 administration led to a significant elevation in MBP transcription in brain biopsies of treated mice. These findings collectively suggest a crucial role for IL-22 in the pathophysiology of MS, particularly in supporting the process of remyelination. These results offer potential avenues for expanding therapeutic strategies for MS treatment. Ongoing experiments aim to further unravel the underlying mechanisms of IL-22 action.
Collapse
Affiliation(s)
- Imen Zamali
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
- Laboratory of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia
- Faculté de Médecine de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Ines Elbini
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Raja Rekik
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Nour-Elhouda Neili
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Wafa Ben Hamouda
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Ahlem Ben Hmid
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
- Laboratory of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia
- Faculté de Médecine de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Raoudha Doghri
- Faculté de Médecine de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Research Laboratory of Precision Medicine, Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
| | - Mélika Ben Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
- Laboratory of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia
- Faculté de Médecine de Tunis, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
3
|
Belghith M, Maghrebi O, Ben Laamari R, Hanachi M, Hrir S, Saied Z, Belal S, Driss A, Ben Sassi S, Boussoffara T, Barbouche MR. Increased IL-22 in cerebrospinal fluid of neuro-behçet's disease patients. Cytokine 2024; 179:156617. [PMID: 38631183 DOI: 10.1016/j.cyto.2024.156617] [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: 09/10/2023] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
Remitting-Relapsing Multiple Sclerosis (RRMS) and Neuro-Behçet Disease (NBD) are two chronic neuro-inflammatory disorders leading to brain damage and disability in young adults. Herein, we investigated in these patients the cytokine response by beads-based multiplex assays during the early stages of these disorders. Cytokine investigations were carried out on treatment-naive patients suffering from RRMS and NBD recruited at the first episode of clinical relapse. Our findings demonstrate that Cerebrospinal Fluid (CSF) cells from NBD patients, but not RRMS, secrete significant high levels of IL-22 which is associated with elevated IL-22 mRNA expression. We also observed an increase in IL-22 levels in the definite NBD subgroup as compared to the probable NBD one, indicating a clear relationship between elevated IL-22 levels and diagnostic certainty. Interestingly, we found no correlation of IL-22 secretion between CSF and serum arguing about intrathecal release of IL-22 in the CNS of NBD patients. Moreover, we showed by correlogram analysis that this cytokine doesn't correlate with IL-17A, IL-17F and IL-21 suggesting that this cytokine is secreted by Th22 cells and not by Th17 cells in the CSF of NBD patients. Finally, we found elevated levels of IL-6 and a positive correlation between IL and 6 and IL-22 in the CSF of NBD. In conclusion, these results suggest that IL-6 contributes to the production of IL-22 by T cells leading to the exacerbation of inflammation and damage within the CNS of NBD patients.
Collapse
Affiliation(s)
- Meriam Belghith
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia.
| | - Olfa Maghrebi
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Faculty of Medicine of Tunis, 1006, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia
| | - Rafika Ben Laamari
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia
| | - Mariem Hanachi
- Laboratory of Bioinformatics, Biomathematics and Biostatistics-LR16IPT09, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia
| | - Sana Hrir
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia
| | - Zakaria Saied
- Faculty of Medicine of Tunis, 1006, Tunisia; Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Samir Belal
- Faculty of Medicine of Tunis, 1006, Tunisia; Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Adel Driss
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA.
| | - Samia Ben Sassi
- Faculty of Medicine of Tunis, 1006, Tunisia; Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Thouraya Boussoffara
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia.
| | - Mohamed-Ridha Barbouche
- Department of Microbiology, Immunology and Infectious Diseases, College of Medicine and Medical Science, Arabian Gulf University 26671, Manama, Bahrain.
| |
Collapse
|
4
|
Kim WJ, Ryu R, Doo EH, Choi Y, Kim K, Kim BK, Kim H, Kim M, Huh CS. Supplementation with the Probiotic Strains Bifidobacterium longum and Lactiplantibacillus rhamnosus Alleviates Glucose Intolerance by Restoring the IL-22 Response and Pancreatic Beta Cell Dysfunction in Type 2 Diabetic Mice. Probiotics Antimicrob Proteins 2023:10.1007/s12602-023-10156-5. [PMID: 37804432 DOI: 10.1007/s12602-023-10156-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 10/09/2023]
Abstract
Type 2 diabetes (T2D) is known as adult-onset diabetes, but recently, T2D has increased in the number of younger people, becoming a major clinical burden in human society. The objective of this study was to determine the effects of Bifidobacterium and Lactiplantibacillus strains derived from the feces of 20 healthy humans on T2D development and to understand the mechanism underlying any positive effects of probiotics. We found that Bifidobacterium longum NBM7-1 (Chong Kun Dang strain 1; CKD1) and Lactiplantibacillus rhamnosus NBM17-4 (Chong Kun Dang strain 2; CKD2) isolated from the feces of healthy Korean adults (n = 20) have anti-diabetic effects based on the insulin sensitivity. During the oral gavage for 8 weeks, T2D mice were supplemented with anti-diabetic drugs (1.0-10 mg/kg body weight) to four positive and negative control groups or four probiotics (200 uL; 1 × 109 CFU/mL) to groups separately or combined to the four treatment groups (n = 6 per group). While acknowledging the relatively small sample size, this study provides valuable insights into the potential benefits of B. longum NBM7-1 and L. rhamnosus NBM17-4 in mitigating T2D development. The animal gene expression was assessed using a qRT-PCR, and metabolic parameters were assessed using an ELISA assay. We demonstrated that B. longum NBM7-1 in the CKD1 group and L. rhamnosus NBM17-4 in the CKD2 group alleviate T2D development through the upregulation of IL-22, which enhances insulin sensitivity and pancreatic functions while reducing liver steatosis. These findings suggest that B. longum NBM7-1 and L. rhamnosus NBM17-4 could be the candidate probiotics for the therapeutic treatments of T2D patients as well as the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Won Jun Kim
- Department of Agricultural Biotechnology, College of Agriculture Sciences, Seoul National University, Seoul, South Korea
| | - Ri Ryu
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, South Korea
| | - Eun-Hee Doo
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, South Korea
- Department of Yuhan Biotechnology, School of Bio-Health Sciences, Yuhan University, Bucheon, 14780, South Korea
| | - Yukyung Choi
- Research Institute, Chong Kun Dang Bio Co. Ltd, Ansan, South Korea
| | - Kyunghwan Kim
- Research Institute, Chong Kun Dang Bio Co. Ltd, Ansan, South Korea
| | - Byoung Kook Kim
- Research Institute, Chong Kun Dang Bio Co. Ltd, Ansan, South Korea
| | - Heebal Kim
- Department of Agricultural Biotechnology, College of Agriculture Sciences, Seoul National University, Seoul, South Korea
- Department of Animal Science and Biotechnology, Seoul National University, Seoul, South Korea
| | - Myunghoo Kim
- Department of Animal Science, Pusan National University, Miryang, South Korea.
| | - Chul Sung Huh
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, South Korea.
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, South Korea.
| |
Collapse
|
5
|
Butyrate promotes post-stroke outcomes in aged mice via interleukin-22. Exp Neurol 2023; 363:114351. [PMID: 36804553 DOI: 10.1016/j.expneurol.2023.114351] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Aging increases the risk of stroke, may exacerbate neuroinflammatory responses, reduce angiogenesis, and promote white matter damage post-stroke, all of which contribute to long-term functional recovery. Butyric acid, an important gut microbial metabolite, showed the highest correlation with the outcomes of ischemic stroke, and butyrate was selected as an effective treatment for aged stroke mice. Here, we tested the neurorestorative effect and potential therapeutic mechanisms of butyrate in aged mice with stroke. Aged male C57BL/6 J mice (17-19 months) were subjected to photothrombotic stroke. We performed butyrate supplementation in the drinking water for 3 weeks before surgery until 14 days after the stroke. At 14 days after ischemic stroke, white matter damage, leukocyte infiltration, and blood-brain barrier permeability were all decreased in the aged stroke mice that received the butyrate treatment, which also improved neurological outcomes by stimulating angiogenesis. Stroke reduces the level of interleukin-22 (IL-22) and butyrate treatment significantly enhanced IL-22 expression in the brain. To further validate the mechanisms of butyrate promoting neurological function after stroke, monoclonal antibodies were used to block IL-22 in aged stroke mice when butyrate treatment was provided. Blocking IL-22 in butyrate-treated aged stroke fails to improve functional outcomes and attenuated butyrate-induced angiogenesis, increased axon/white matter density and blood-brain barrier (BBB) integrity, but has no effect on inflammatory cells infiltration. In conclusion, butyrate improves outcomes in aged mice after stroke by promoting angiogenesis and BBB integrity and reducing leukocyte infiltration. To some extent, IL-22 may contribute to butyrate treatment induced vascular remodeling and increased BBB integrity responses in aged stroke mice.
Collapse
|
6
|
Abstract
Inflammation is a biological process that dynamically alters the surrounding microenvironment, including participating immune cells. As a well-protected organ surrounded by specialized barriers and with immune privilege properties, the central nervous system (CNS) tightly regulates immune responses. Yet in neuroinflammatory conditions, pathogenic immunity can disrupt CNS structure and function. T cells in particular play a key role in promoting and restricting neuroinflammatory responses, while the inflamed CNS microenvironment can influence and reshape T cell function and identity. Still, the contraction of aberrant T cell responses within the CNS is not well understood. Using autoimmunity as a model, here we address the contribution of CD4 T helper (Th) cell subsets in promoting neuropathology and disease. To address the mechanisms antagonizing neuroinflammation, we focus on the control of the immune response by regulatory T cells (Tregs) and describe the counteracting processes that preserve their identity under inflammatory challenges. Finally, given the influence of the local microenvironment on immune regulation, we address how CNS-intrinsic signals reshape T cell function to mitigate abnormal immune T cell responses.
Collapse
Affiliation(s)
- Nail Benallegue
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000, Nantes, France
| | - Hania Kebir
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jorge I. Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
7
|
Eken A, Erdem S, Haliloglu Y, Zehra Okus F, Cakir M, Fatih Yetkin M, Akcakoyunlu M, Karayigit MO, Azizoglu ZB, Bicer A, Gur TN, Aslan K, Hora M, Oukka M, Altuntas HD, Ufuk Nalbantoglu O, Gundogdu A, Mirza M, Canatan H. Temporal overexpression of IL-22 and Reg3γ differentially impacts the severity of experimental autoimmune encephalomyelitis. Immunology 2021; 164:73-89. [PMID: 33876425 PMCID: PMC8358722 DOI: 10.1111/imm.13340] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022] Open
Abstract
IL-22 is an alpha-helical cytokine which belongs to the IL-10 family of cytokines. IL-22 is produced by RORγt+ innate and adaptive lymphocytes, including ILC3, γδ T, iNKT, Th17 and Th22 cells and some granulocytes. IL-22 receptor is expressed primarily by non-haematopoietic cells. IL-22 is critical for barrier immunity at the mucosal surfaces in the steady state and during infection. Although IL-22 knockout mice were previously shown to develop experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS), how temporal IL-22 manipulation in adult mice would affect EAE course has not been studied previously. In this study, we overexpressed IL-22 via hydrodynamic gene delivery or blocked it via neutralizing antibodies in C57BL/6 mice to explore the therapeutic impact of IL-22 modulation on the EAE course. IL-22 overexpression significantly decreased EAE scores and demyelination, and reduced infiltration of IFN-γ+IL-17A+Th17 cells into the central nervous system (CNS). The neutralization of IL-22 did not alter the EAE pathology significantly. We show that IL-22-mediated protection is independent of Reg3γ, an epithelial cell-derived antimicrobial peptide induced by IL-22. Thus, overexpression of Reg3γ significantly exacerbated EAE scores, demyelination and infiltration of IFN-γ+IL-17A+ and IL-17A+GM-CSF+Th17 cells to CNS. We also show that Reg3γ may inhibit IL-2-mediated STAT5 signalling and impair expansion of Treg cells in vivo and in vitro. Finally, Reg3γ overexpression dramatically impacted intestinal microbiota during EAE. Our results provide novel insight into the role of IL-22 and IL-22-induced antimicrobial peptide Reg3γ in the pathogenesis of CNS inflammation in a murine model of MS.
Collapse
Affiliation(s)
- Ahmet Eken
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Serife Erdem
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Yesim Haliloglu
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Fatma Zehra Okus
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Mustafa Cakir
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
- Department of Medical BiologyVan Yuzuncu Yıl University School of MedicineVanTurkey
| | | | - Merve Akcakoyunlu
- Department of NeurologyErciyes University School of MedicineKayseriTurkey
| | | | - Zehra Busra Azizoglu
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Ayten Bicer
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Tugba Nur Gur
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Kubra Aslan
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Mehmet Hora
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Mohamed Oukka
- Department of ImmunologyUniversity of WashingtonSeattleWAUSA
| | - Hamiyet Donmez Altuntas
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| | - Ozkan Ufuk Nalbantoglu
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
- Department of Computer EngineeringFaculty of EngineeringErciyes UniversityKayseriTurkey
| | - Aycan Gundogdu
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
- Department of Microbiology and Clinical MicrobiologyErciyes University School of MedicineKayseriTurkey
| | - Meral Mirza
- Department of NeurologyErciyes University School of MedicineKayseriTurkey
| | - Halit Canatan
- Department of Medical BiologyErciyes University School of MedicineKayseriTurkey
- Betül‐Ziya Eren Genome and Stem Cell Center (GENKOK)KayseriTurkey
| |
Collapse
|
8
|
Jiang Q, Yang G, Xiao F, Xie J, Wang S, Lu L, Cui D. Role of Th22 Cells in the Pathogenesis of Autoimmune Diseases. Front Immunol 2021; 12:688066. [PMID: 34295334 PMCID: PMC8290841 DOI: 10.3389/fimmu.2021.688066] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Upon antigenic stimulation, naïve CD4+T cells differentiate into different subsets and secrete various cytokines to exert biological effects. Th22 cells, a newly identified CD4+T cell subset,are distinct from the Th1, Th2 and Th17 subsets. Th22 cells secrete certain cytokines such as IL-22, IL-13 and TNF-α, but not others, such as IL-17, IL-4, or interferon-γ (IFN-γ), and they express chemokine receptors CCR4, CCR6 and CCR10. Th22 cells were initially found to play a role in skin inflammatory diseases, but recent studies have demonstrated their involvement in the development of various autoimmune diseases. Here, we review research advances in the origin, characteristics and effector mechanisms of Th22 cells, with an emphasis on the role of Th22 cells and their main effector cytokine IL-22 in the pathogenesis of autoimmune diseases. The findings presented here may facilitate the development of new therapeutic strategies for targeting these diseases.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Blood Transfusion, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Guocan Yang
- Department of Blood Transfusion, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Fan Xiao
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong; Chongqing International Institute for Immunology, Chongqing, China
| | - Jue Xie
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong; Chongqing International Institute for Immunology, Chongqing, China
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Lindborg JA, Tran NM, Chenette DM, DeLuca K, Foli Y, Kannan R, Sekine Y, Wang X, Wollan M, Kim IJ, Sanes JR, Strittmatter SM. Optic nerve regeneration screen identifies multiple genes restricting adult neural repair. Cell Rep 2021; 34:108777. [PMID: 33657370 PMCID: PMC8009559 DOI: 10.1016/j.celrep.2021.108777] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/21/2020] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Adult mammalian central nervous system (CNS) trauma interrupts neural networks and, because axonal regeneration is minimal, neurological deficits persist. Repair via axonal growth is limited by extracellular inhibitors and cell-autonomous factors. Based on results from a screen in vitro, we evaluate nearly 400 genes through a large-scale in vivo regeneration screen. Suppression of 40 genes using viral-driven short hairpin RNAs (shRNAs) promotes retinal ganglion cell (RGC) axon regeneration after optic nerve crush (ONC), and most are validated by separate CRISPR-Cas9 editing experiments. Expression of these axon-regeneration-suppressing genes is not significantly altered by axotomy. Among regeneration-limiting genes, loss of the interleukin 22 (IL-22) cytokine allows an early, yet transient, inflammatory response in the retina after injury. Reduced IL-22 drives concurrent activation of signal transducer and activator of transcription 3 (Stat3) and dual leucine zipper kinase (DLK) pathways and upregulation of multiple neuron-intrinsic regeneration-associated genes (RAGs). Including IL-22, our screen identifies dozens of genes that limit CNS regeneration. Suppression of these genes in the context of axonal damage could support improved neural repair.
Collapse
Affiliation(s)
- Jane A Lindborg
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Nicholas M Tran
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Devon M Chenette
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Kristin DeLuca
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Yram Foli
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ramakrishnan Kannan
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Yuichi Sekine
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Xingxing Wang
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Marius Wollan
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - In-Jung Kim
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
10
|
Abstract
Interleukin-22 (IL-22) is secreted by a wide range of immune cells and its downstream effects are mediated by the IL-22 receptor, which is present on non-immune cells in many organs throughout the body. IL-22 is an inflammatory mediator that conditions the tissue compartment by upregulating innate immune responses and is also a homeostatic factor that promotes tissue integrity and regeneration. Interestingly, the IL-22 system has also been linked to many T cell driven inflammatory diseases. Despite this, the downstream effects of IL-22 on the adaptive immune system has received little attention. We have reviewed the literature for experimental data that suggest IL-22 mediated effects on T cells, either transduced directly or via mediators expressed by innate immune cells or non-immune cells in response to IL-22. Collectively, the reviewed data indicate that IL-22 has a hitherto unappreciated influence on T helper cell polarization, or the secretion of signature cytokines, that is context dependent but in many cases results in a reduction of the Th1 type response and to some extent promotion of regulatory T cells. Further studies are needed that specifically address these aspects of IL-22 signaling, which can benefit the understanding and treatment of a wide range of diseases.
Collapse
Affiliation(s)
- Hannes Lindahl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
11
|
Kunkl M, Frascolla S, Amormino C, Volpe E, Tuosto L. T Helper Cells: The Modulators of Inflammation in Multiple Sclerosis. Cells 2020; 9:cells9020482. [PMID: 32093011 PMCID: PMC7072830 DOI: 10.3390/cells9020482] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by the progressive loss of axonal myelin in several areas of the central nervous system (CNS) that is responsible for clinical symptoms such as muscle spasms, optic neuritis, and paralysis. The progress made in more than one decade of research in animal models of MS for clarifying the pathophysiology of MS disease validated the concept that MS is an autoimmune inflammatory disorder caused by the recruitment in the CNS of self-reactive lymphocytes, mainly CD4+ T cells. Indeed, high levels of T helper (Th) cells and related cytokines and chemokines have been found in CNS lesions and in cerebrospinal fluid (CSF) of MS patients, thus contributing to the breakdown of the blood-brain barrier (BBB), the activation of resident astrocytes and microglia, and finally the outcome of neuroinflammation. To date, several types of Th cells have been discovered and designated according to the secreted lineage-defining cytokines. Interestingly, Th1, Th17, Th1-like Th17, Th9, and Th22 have been associated with MS. In this review, we discuss the role and interplay of different Th cell subpopulations and their lineage-defining cytokines in modulating the inflammatory responses in MS and the approved as well as the novel therapeutic approaches targeting T lymphocytes in the treatment of the disease.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Simone Frascolla
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Elisabetta Volpe
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| |
Collapse
|
12
|
Gómez-Fernández P, Lopez de Lapuente Portilla A, Astobiza I, Mena J, Urtasun A, Altmann V, Matesanz F, Otaegui D, Urcelay E, Antigüedad A, Malhotra S, Montalban X, Castillo-Triviño T, Espino-Paisán L, Aktas O, Buttmann M, Chan A, Fontaine B, Gourraud PA, Hecker M, Hoffjan S, Kubisch C, Kümpfel T, Luessi F, Zettl UK, Zipp F, Alloza I, Comabella M, Lill CM, Vandenbroeck K. The Rare IL22RA2 Signal Peptide Coding Variant rs28385692 Decreases Secretion of IL-22BP Isoform-1, -2 and -3 and Is Associated with Risk for Multiple Sclerosis. Cells 2020; 9:cells9010175. [PMID: 31936765 PMCID: PMC7017210 DOI: 10.3390/cells9010175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 12/29/2019] [Accepted: 01/03/2020] [Indexed: 10/29/2022] Open
Abstract
The IL22RA2 locus is associated with risk for multiple sclerosis (MS) but causative variants are yet to be determined. In a single nucleotide polymorphism (SNP) screen of this locus in a Basque population, rs28385692, a rare coding variant substituting Leu for Pro at position 16 emerged significantly (p = 0.02). This variant is located in the signal peptide (SP) shared by the three secreted protein isoforms produced by IL22RA2 (IL-22 binding protein-1(IL-22BPi1), IL-22BPi2 and IL-22BPi3). Genotyping was extended to a Europe-wide case-control dataset and yielded high significance in the full dataset (p = 3.17 × 10-4). Importantly, logistic regression analyses conditioning on the main known MS-associated SNP at this locus, rs17066096, revealed that this association was independent from the primary association signal in the full case-control dataset. In silico analysis predicted both disruption of the alpha helix of the H-region of the SP and decreased hydrophobicity of this region, ultimately affecting the SP cleavage site. We tested the effect of the p.Leu16Pro variant on the secretion of IL-22BPi1, IL-22BPi2 and IL-22BPi3 and observed that the Pro16 risk allele significantly lowers secretion levels of each of the isoforms to around 50%-60% in comparison to the Leu16 reference allele. Thus, our study suggests that genetically coded decreased levels of IL-22BP isoforms are associated with augmented risk for MS.
Collapse
Affiliation(s)
- Paloma Gómez-Fernández
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (P.G.-F.); (A.L.d.L.P.); (I.A.); (J.M.); (A.U.); (I.A.)
| | - Aitzkoa Lopez de Lapuente Portilla
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (P.G.-F.); (A.L.d.L.P.); (I.A.); (J.M.); (A.U.); (I.A.)
- Department of Laboratory Medicine, Lund University, SE-221 00 Lund, Sweden
| | - Ianire Astobiza
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (P.G.-F.); (A.L.d.L.P.); (I.A.); (J.M.); (A.U.); (I.A.)
| | - Jorge Mena
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (P.G.-F.); (A.L.d.L.P.); (I.A.); (J.M.); (A.U.); (I.A.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Andoni Urtasun
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (P.G.-F.); (A.L.d.L.P.); (I.A.); (J.M.); (A.U.); (I.A.)
| | - Vivian Altmann
- Genetic and Molecular Epidemiology Group, Lübeck Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, 23552 Lübeck, Germany; (V.A.); (C.M.L.)
| | - Fuencisla Matesanz
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina López Neyra (IPBLN), CSIC, 18002 Granada, Spain;
| | - David Otaegui
- Multiple Sclerosis Group, Biodonostia Research Institute, Paseo Doctor Begiristain, s/n, 20014 San Sebastián, Spain; (D.O.); (T.C.-T.)
| | - Elena Urcelay
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, 28014 Madrid, Spain; (E.U.); (L.E.-P.)
| | | | - Sunny Malhotra
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08007 Barcelona, Spain; (S.M.); (X.M.); (M.C.)
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08007 Barcelona, Spain; (S.M.); (X.M.); (M.C.)
| | - Tamara Castillo-Triviño
- Multiple Sclerosis Group, Biodonostia Research Institute, Paseo Doctor Begiristain, s/n, 20014 San Sebastián, Spain; (D.O.); (T.C.-T.)
| | - Laura Espino-Paisán
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, 28014 Madrid, Spain; (E.U.); (L.E.-P.)
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Mathias Buttmann
- Department of Neurology, University of Wuerzburg, 97080 Wuerzburg, Germany;
- Department of Neurology, Caritas Hospital, 97980 Bad Mergentheim, Germany
| | - Andrew Chan
- Department of Neurology, Inselspital Bern, Bern University Hospital, University of Bern, 3011 Bern, Switzerland;
| | - Bertrand Fontaine
- INSERM, Sorbonne University, Assistance Publique-Hopitaux de Paris (AP-HP), UMR 974 and Neuro-Myology Service, University Hospital Pitié-Salpêtrière, 75013 Paris, France;
| | - Pierre-Antoine Gourraud
- Nantes Université, CHU, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ATIP-Avenir, Equipe 5, 44093 Nantes, France;
- CHU de Nantes, INSERM, CIC 1413, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des données, 44000 Nantes, France
| | - Michael Hecker
- Department of Neurology, Neuroimmunological Section, University of Rostock, 18147 Rostock, Germany; (M.H.); (U.K.Z.)
| | - Sabine Hoffjan
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians University, 80333 Munich, Germany;
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, 55116 Mainz, Germany; (F.L.); (F.Z.)
| | - Uwe K. Zettl
- Department of Neurology, Neuroimmunological Section, University of Rostock, 18147 Rostock, Germany; (M.H.); (U.K.Z.)
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, 55116 Mainz, Germany; (F.L.); (F.Z.)
| | - Iraide Alloza
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (P.G.-F.); (A.L.d.L.P.); (I.A.); (J.M.); (A.U.); (I.A.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08007 Barcelona, Spain; (S.M.); (X.M.); (M.C.)
| | - Christina M. Lill
- Genetic and Molecular Epidemiology Group, Lübeck Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, 23552 Lübeck, Germany; (V.A.); (C.M.L.)
- Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, 55116 Mainz, Germany; (F.L.); (F.Z.)
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, 23552 Lübeck, Germany
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College, London SW71, UK
| | - Koen Vandenbroeck
- Neurogenomiks Laboratory, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (P.G.-F.); (A.L.d.L.P.); (I.A.); (J.M.); (A.U.); (I.A.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
- Correspondence: ; Tel.: +34-946182622 (ext. 844748)
| |
Collapse
|
13
|
Tahrali I, Kucuksezer UC, Akdeniz N, Altintas A, Uygunoglu U, Aktas-Cetin E, Deniz G. CD3 -CD56 + NK cells display an inflammatory profile in RR-MS patients. Immunol Lett 2019; 216:63-69. [PMID: 31589897 DOI: 10.1016/j.imlet.2019.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/22/2019] [Accepted: 10/03/2019] [Indexed: 01/06/2023]
Abstract
Multiple Sclerosis (MS) is an immune-mediated and neurodegenerative disease of central nervous system. Relapsing-remitting (RR)-MS occurring with acute attacks and remissions, is the most common clinical type of MS. There are different strategies applied in first-line treatment of RR-MS patients such as interferon-beta (IFN-β) and glatiramer acetate. In this study, activating and inhibitory receptor expressions and interleukin (IL)-22 levels of NK cells were investigated in RR-MS patients with or without IFN-β therapy. Activating receptor expression and IL-22 levels of NK cells were increased in RR-MS patients under IFN-β therapy. Elevated NK cells with activating profile and increased IL-22 under IFN-β therapy suggest that IFN-β treatment might direct NK cells toward a pro-inflammatory status.
Collapse
Affiliation(s)
- Ilhan Tahrali
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Umut Can Kucuksezer
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Nilgun Akdeniz
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Ayse Altintas
- Koc University, Faculty of Medicine, Department of Neurology, Istanbul, Turkey; Istanbul University Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Ugur Uygunoglu
- Istanbul University Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Esin Aktas-Cetin
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | - Gunnur Deniz
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey.
| |
Collapse
|
14
|
Abood RN, McHugh KJ, Rich HE, Ortiz MA, Tobin JM, Ramanan K, Robinson KM, Bomberger JM, Kolls JK, Manni ML, Pociask DA, Alcorn JF. IL-22-binding protein exacerbates influenza, bacterial super-infection. Mucosal Immunol 2019; 12:1231-1243. [PMID: 31296910 PMCID: PMC6717528 DOI: 10.1038/s41385-019-0188-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 06/15/2019] [Accepted: 06/24/2019] [Indexed: 02/04/2023]
Abstract
Secondary bacterial pneumonia is a significant complication of severe influenza infection and Staphylococcus aureus and Streptococcus pneumoniae are the primary pathogens of interest. IL-22 promotes S. aureus and S. pneumoniae host defense in the lung through epithelial integrity and induction of antimicrobial peptides and is inhibited by the soluble decoy receptor IL-22-binding protein (IL-22BP). Little is known about the effect of the IL-22/IL-22BP regulatory pathway on lung infection, and it has not been studied in the setting of super-infection. We exposed wild-type and IL-22BP-/- mice to influenza A/PR/8/34 for 6 days prior to infection with S. aureus (USA300) S. pneumoniae. Super-infected IL-22BP-/- mice had decreased bacterial burden and improved survival compared to controls. IL-22BP-/- mice exhibited decreased inflammation, increased lipocalin 2 expression, and deletion of IL-22BP was associated with preserved epithelial barrier function with evidence of improved tight junction stability. Human bronchial epithelial cells treated with IL-22Fc showed evidence of improved tight junctions compared to untreated cells. This study revealed that IL-22BP-/- mice are protected during influenza, bacterial super-infection, suggesting that IL-22BP has a pro-inflammatory role and impairs epithelial barrier function likely through interaction with IL-22.
Collapse
Affiliation(s)
- Robert N Abood
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin J McHugh
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Helen E Rich
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Marianna A Ortiz
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Joshua M Tobin
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Krishnaveni Ramanan
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Keven M Robinson
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jennifer M Bomberger
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Michelle L Manni
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Derek A Pociask
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - John F Alcorn
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Waubant E, Lucas R, Mowry E, Graves J, Olsson T, Alfredsson L, Langer‐Gould A. Environmental and genetic risk factors for MS: an integrated review. Ann Clin Transl Neurol 2019; 6:1905-1922. [PMID: 31392849 PMCID: PMC6764632 DOI: 10.1002/acn3.50862] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/11/2022] Open
Abstract
Recent findings have provided a molecular basis for the combined contributions of multifaceted risk factors for the onset of multiple sclerosis (MS). MS appears to start as a chronic dysregulation of immune homeostasis resulting from complex interactions between genetic predispositions, infectious exposures, and factors that lead to pro-inflammatory states, including smoking, obesity, and low sun exposure. This is supported by the discovery of gene-environment (GxE) interactions and epigenetic alterations triggered by environmental exposures in individuals with particular genetic make-ups. It is notable that several of these pro-inflammatory factors have not emerged as strong prognostic indicators. Biological processes at play during the relapsing phase of the disease may result from initial inflammatory-mediated injury, while risk factors for the later phase of MS, which is weighted toward neurodegeneration, are not yet well defined. This integrated review of current evidence guides recommendations for clinical practice and highlights research gaps.
Collapse
Affiliation(s)
| | - Robyn Lucas
- National Centre for Epidemiology and Population Health, Research School of Population HealthAustralian National UniversityCanberraAustralia
| | - Ellen Mowry
- Department of Neurology and EpidemiologyJohns Hopkins UniversityBaltimoreMaryland
| | | | - Tomas Olsson
- Department of NeurologyKarolinska Institutet, Department of Clinical NeuroscienceStockholmSweden
| | - Lars Alfredsson
- Department of EpidemiologyInstitute of Environmental Medicine, Karolinska InstitutetStockholmSweden
| | - Annette Langer‐Gould
- Clinical & Translational NeuroscienceKaiser Permanente/Southern California Permanente Medical GroupLos AngelesCalifornia
| |
Collapse
|
16
|
Zarobkiewicz MK, Kowalska W, Slawinski M, Rolinski J, Bojarska-Junak A. The role of interleukin 22 in multiple sclerosis and its association with c-Maf and AHR. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2019; 163:200-206. [PMID: 31162488 DOI: 10.5507/bp.2019.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023] Open
Abstract
The aim of this paper was to summarise knowledge of IL-22 involvement in multiple sclerosis (MS) and the possible link between IL-22 and two transcription factors - AHR and c-Maf. The conclusion is that despite numerous studies, the exact role of IL-22 in the pathogenesis of MS is still unknown. The expression and function of c-Maf in MS have not been studied. It seems that the functions of c-Maf and AHR are at least partly connected with IL-22, as both directly or indirectly influence the regulation of IL-22 expression. This possible connection has never been studied in MS.
Collapse
Affiliation(s)
| | - Wioleta Kowalska
- Chair and Department of Clinical Immunology, Medical University of Lublin, Lublin, Poland
| | - Miroslaw Slawinski
- Chair and Department of Histology and Embryology with Experimental Cytology Unit, Medical University of Lublin, Lublin, Poland
| | - Jacek Rolinski
- Chair and Department of Clinical Immunology, Medical University of Lublin, Lublin, Poland
| | | |
Collapse
|
17
|
Mattapallil MJ, Kielczewski JL, Zárate-Bladés CR, St Leger AJ, Raychaudhuri K, Silver PB, Jittayasothorn Y, Chan CC, Caspi RR. Interleukin 22 ameliorates neuropathology and protects from central nervous system autoimmunity. J Autoimmun 2019; 102:65-76. [PMID: 31080013 PMCID: PMC6667188 DOI: 10.1016/j.jaut.2019.04.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/01/2019] [Accepted: 04/18/2019] [Indexed: 01/07/2023]
Abstract
IL-22 has opposing effects in different tissues, from pro-inflammatory (skin, joints) to protective (liver, intestine) but little is known about its effects on neuroinflammation. We examined the effect of IL-22 on retinal tissue by using the model of experimental autoimmune uveitis (EAU) in IL-22-/- mice, as well as by intraocular injections of recombinant IL-22 or anti-IL-22 antibodies in wild type animals. During EAU, IL-22 was produced in the eye by CD4+ eye-infiltrating T cells. EAU-challenged IL-22-/- mice, as well as WT mice treated systemically or intraocularly with anti-IL-22 antibodies during the expression phase of disease, developed exacerbated retinal damage. Furthermore, IL-22-/- mice were more susceptible than WT controls to glutamate-induced neurotoxicity, whereas local IL-22 supplementation was protective, suggesting direct or indirect neuroprotective effects. Mechanistic studies revealed that retinal glial Müller cells express IL-22rα1 in vivo, and in vitro IL-22 enhanced their ability to suppress proliferation of effector T cells. Finally, IL-22 injected into the eye concurrently with IL-1, inhibited the (IL-1-induced) expression of multiple proinflammatory and proapoptotic genes in retinal tissue. These findings suggest that IL-22 can function locally within the retina to reduce inflammatory damage and provide neuroprotection by affecting multiple molecular and cellular pathways.
Collapse
Affiliation(s)
- Mary J Mattapallil
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jennifer L Kielczewski
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Carlos R Zárate-Bladés
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anthony J St Leger
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kumarkrishna Raychaudhuri
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Phyllis B Silver
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yingyos Jittayasothorn
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chi-Chao Chan
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
18
|
Lindahl H, Guerreiro-Cacais AO, Bedri SK, Linnerbauer M, Lindén M, Abdelmagid N, Tandre K, Hollins C, Irving L, Glover C, Jones C, Alfredsson L, Rönnblom L, Kockum I, Khademi M, Jagodic M, Olsson T. IL-22 Binding Protein Promotes the Disease Process in Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2019; 203:888-898. [PMID: 31292217 DOI: 10.4049/jimmunol.1900400] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/18/2019] [Indexed: 12/14/2022]
Abstract
Genome-wide association studies have mapped the specific sequence variants that predispose for multiple sclerosis (MS). The pathogenic mechanisms that underlie these associations could be leveraged to develop safer and more effective MS treatments but are still poorly understood. In this article, we study the genetic risk variant rs17066096 and the candidate gene that encodes IL-22 binding protein (IL-22BP), an antagonist molecule of the cytokine IL-22. We show that monocytes from carriers of the risk genotype of rs17066096 express more IL-22BP in vitro and cerebrospinal fluid levels of IL-22BP correlate with MS lesion load on magnetic resonance imaging. We confirm the pathogenicity of IL-22BP in both rat and mouse models of MS and go on to suggest a pathogenic mechanism involving lack of IL-22-mediated inhibition of T cell-derived IFN-γ expression. Our results demonstrate a pathogenic role of IL-22BP in three species with a potential mechanism of action involving T cell polarization, suggesting a therapeutic potential of IL-22 in the context of MS.
Collapse
Affiliation(s)
- Hannes Lindahl
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - André O Guerreiro-Cacais
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Sahl Khalid Bedri
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Mathias Linnerbauer
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Magdalena Lindén
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Nada Abdelmagid
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Karolina Tandre
- Department of Medical Sciences, Rheumatology and Science for Life Laboratories, Uppsala University, 75105 Uppsala, Sweden
| | - Claire Hollins
- Respiratory, Inflammation and Autoimmunity Research, MedImmune, Cambridge CB21 6GH, United Kingdom
| | - Lorraine Irving
- High Content Imaging, MedImmune, Cambridge CB21 6GH, United Kingdom; and
| | - Colin Glover
- Respiratory, Inflammation and Autoimmunity Research, MedImmune, Cambridge CB21 6GH, United Kingdom
| | - Clare Jones
- Respiratory, Inflammation and Autoimmunity Research, MedImmune, Cambridge CB21 6GH, United Kingdom
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology and Science for Life Laboratories, Uppsala University, 75105 Uppsala, Sweden
| | - Ingrid Kockum
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Mohsen Khademi
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Maja Jagodic
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
19
|
Gómez-Fernández P, Urtasun A, Astobiza I, Mena J, Alloza I, Vandenbroeck K. Pharmacological Targeting of the ER-Resident Chaperones GRP94 or Cyclophilin B Induces Secretion of IL-22 Binding Protein Isoform-1 (IL-22BPi1). Int J Mol Sci 2019; 20:ijms20102440. [PMID: 31108847 PMCID: PMC6566634 DOI: 10.3390/ijms20102440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 01/05/2023] Open
Abstract
Of the three interleukin-22 binding protein (IL-22BP) isoforms produced by the human IL22RA2 gene, IL-22BPi2 and IL-22BPi3 are capable of neutralizing IL-22. The longest isoform, IL-22BPi1, does not bind IL-22, is poorly secreted, and its retention within the endoplasmic reticulum (ER) is associated with induction of an unfolded protein response (UPR). Therapeutic modulation of IL-22BPi2 and IL-22BPi3 production may be beneficial in IL-22-dependent disorders. Recently, we identified the ER chaperones GRP94 and cyclophilin B in the interactomes of both IL-22BPi1 and IL-22BPi2. In this study, we investigated whether secretion of the IL-22BP isoforms could be modulated by pharmacological targeting of GRP94 and cyclophilin B, either by means of geldanamycin, that binds to the ADP/ATP pocket shared by HSP90 paralogs, or by cyclosporin A, which causes depletion of ER cyclophilin B levels through secretion. We found that geldanamycin and its analogs did not influence secretion of IL-22BPi2 or IL-22BPi3, but significantly enhanced intracellular and secreted levels of IL-22BPi1. The secreted protein was heterogeneously glycosylated, with both high-mannose and complex-type glycoforms present. In addition, cyclosporine A augmented the secretion of IL-22BPi1 and reduced that of IL-22BPi2 and IL-22BPi3. Our data indicate that the ATPase activity of GRP94 and cyclophilin B are instrumental in ER sequestration and degradation of IL-22BPi1, and that blocking these factors mobilizes IL-22BPi1 toward the secretory route.
Collapse
Affiliation(s)
- Paloma Gómez-Fernández
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Andoni Urtasun
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Ianire Astobiza
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Jorge Mena
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Iraide Alloza
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Koen Vandenbroeck
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
20
|
Gómez-Fernández P, Urtasun A, Paton AW, Paton JC, Borrego F, Dersh D, Argon Y, Alloza I, Vandenbroeck K. Long Interleukin-22 Binding Protein Isoform-1 Is an Intracellular Activator of the Unfolded Protein Response. Front Immunol 2018; 9:2934. [PMID: 30619294 PMCID: PMC6302113 DOI: 10.3389/fimmu.2018.02934] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/29/2018] [Indexed: 12/26/2022] Open
Abstract
The human IL22RA2 gene co-produces three protein isoforms in dendritic cells [IL-22 binding protein isoform-1 (IL-22BPi1), IL-22BPi2, and IL-22BPi3]. Two of these, IL-22BPi2 and IL-22BPi3, are capable of neutralizing the biological activity of IL-22. The function of IL-22BPi1, which differs from IL-22BPi2 through an in-frame 32-amino acid insertion provided by an alternatively spliced exon, remains unknown. Using transfected human cell lines, we demonstrate that IL-22BPi1 is secreted detectably, but at much lower levels than IL-22BPi2, and unlike IL-22BPi2 and IL-22BPi3, is largely retained in the endoplasmic reticulum (ER). As opposed to IL-22BPi2 and IL-22BPi3, IL-22BPi1 is incapable of neutralizing or binding to IL-22 measured in bioassay or assembly-induced IL-22 co-folding assay. We performed interactome analysis to disclose the mechanism underlying the poor secretion of IL-22BPi1 and identified GRP78, GRP94, GRP170, and calnexin as main interactors. Structure-function analysis revealed that, like IL-22BPi2, IL-22BPi1 binds to the substrate-binding domain of GRP78 as well as to the middle domain of GRP94. Ectopic expression of wild-type GRP78 enhanced, and ATPase-defective GRP94 mutant decreased, secretion of both IL-22BPi1 and IL-22BPi2, while neither of both affected IL-22BPi3 secretion. Thus, IL-22BPi1 and IL-22BPi2 are bona fide clients of the ER chaperones GRP78 and GRP94. However, only IL-22BPi1 activates an unfolded protein response (UPR) resulting in increased protein levels of GRP78 and GRP94. Cloning of the IL22RA2 alternatively spliced exon into an unrelated cytokine, IL-2, bestowed similar characteristics on the resulting protein. We also found that CD14++/CD16+ intermediate monocytes produced a higher level of IL22RA2 mRNA than classical and non-classical monocytes, but this difference disappeared in immature dendritic cells (moDC) derived thereof. Upon silencing of IL22RA2 expression in moDC, GRP78 levels were significantly reduced, suggesting that native IL22RA2 expression naturally contributes to upregulating GRP78 levels in these cells. The IL22RA2 alternatively spliced exon was reported to be recruited through a single mutation in the proto-splice site of a Long Terminal Repeat retrotransposon sequence in the ape lineage. Our work suggests that positive selection of IL-22BPi1 was not driven by IL-22 antagonism as in the case of IL-22BPi2 and IL-22BPi3, but by capacity for induction of an UPR response.
Collapse
Affiliation(s)
- Paloma Gómez-Fernández
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Andoni Urtasun
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Adrienne W. Paton
- Research for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - James C. Paton
- Research for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - Francisco Borrego
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Basque Center for Transfusion and Human Tissues, Galdakao, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Devin Dersh
- Division of Cell Pathology, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yair Argon
- Division of Cell Pathology, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Iraide Alloza
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Koen Vandenbroeck
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
21
|
Burmeister AR, Marriott I. The Interleukin-10 Family of Cytokines and Their Role in the CNS. Front Cell Neurosci 2018; 12:458. [PMID: 30542269 PMCID: PMC6277801 DOI: 10.3389/fncel.2018.00458] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022] Open
Abstract
Resident cells of the central nervous system (CNS) play an important role in detecting insults and initiating protective or sometimes detrimental host immunity. At peripheral sites, immune responses follow a biphasic course with the rapid, but transient, production of inflammatory mediators giving way to the delayed release of factors that promote resolution and repair. Within the CNS, it is well known that glial cells contribute to the onset and progression of neuroinflammation, but it is only now becoming apparent that microglia and astrocytes also play an important role in producing and responding to immunosuppressive factors that serve to limit the detrimental effects of such responses. Interleukin-10 (IL-10) is generally considered to be the quintessential immunosuppressive cytokine, and its ability to resolve inflammation and promote wound repair at peripheral sites is well documented. In the present review article, we discuss the evidence for the production of IL-10 by glia, and describe the ability of CNS cells, including microglia and astrocytes, to respond to this suppressive factor. Furthermore, we review the literature for the expression of other members of the IL-10 cytokine family, IL-19, IL-20, IL-22 and IL-24, within the brain, and discuss the evidence of a role for these poorly understood cytokines in the regulation of infectious and sterile neuroinflammation. In concert, the available data indicate that glia can produce IL-10 and the related cytokines IL-19 and IL-24 in a delayed manner, and these cytokines can limit glial inflammatory responses and/or provide protection against CNS insult. However, the roles of other IL-10 family members within the CNS remain unclear, with IL-20 appearing to act as a pro-inflammatory factor, while IL-22 may play a protective role in some instances and a detrimental role in others, perhaps reflecting the pleiotropic nature of this cytokine family. What is clear is that our current understanding of the role of IL-10 and related cytokines within the CNS is limited at best, and further research is required to define the actions of this understudied family in inflammatory brain disorders.
Collapse
Affiliation(s)
- Amanda R Burmeister
- Department of Biological Sciences, The University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Ian Marriott
- Department of Biological Sciences, The University of North Carolina at Charlotte, Charlotte, NC, United States
| |
Collapse
|
22
|
Podbielska M, O'Keeffe J, Hogan EL. Autoimmunity in multiple sclerosis: role of sphingolipids, invariant NKT cells and other immune elements in control of inflammation and neurodegeneration. J Neurol Sci 2017; 385:198-214. [PMID: 29406905 DOI: 10.1016/j.jns.2017.12.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is the most common demyelinating disease of the central nervous system. It is classified as being an autoimmune response in the genetically susceptible individual to a persistent but unidentified antigen(s). Both the adaptive and the innate immune systems are likely to contribute significantly to MS pathogenesis. This review summarizes current understanding of the characteristics of MS autoimmunity in the initiation and progression of the disease. In particular we find it timely to classify the autoimmune responses by focusing on the immunogenic features of myelin-derived lipids in MS including molecular mimicry; on alterations of bioactive sphingolipids mediators in MS; and on functional roles for regulatory effector cells, including innate lymphocyte populations, like the invariant NKT (iNKT) cells which bridge adaptive and innate immune systems. Recent progress in identifying the nature of sphingolipids recognition for iNKT cells in immunity and the functional consequences of the lipid-CD1d interaction opens new avenues of access to the pathogenesis of demyelination in MS as well as design of lipid antigen-specific therapeutics.
Collapse
Affiliation(s)
- Maria Podbielska
- Department of Neurology and Neurosurgery, Medical University of South Carolina Charleston, SC, USA; Laboratory of Signal Transduction Molecules, Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland.
| | - Joan O'Keeffe
- Department of Biopharmaceutical & Medical Science, School of Science & Computing, Galway-Mayo Institute of Technology, Galway, Ireland
| | - Edward L Hogan
- Department of Neurology and Neurosurgery, Medical University of South Carolina Charleston, SC, USA
| |
Collapse
|
23
|
Kulakova O, Bashinskaya V, Kiselev I, Baulina N, Tsareva E, Nikolaev R, Kozin M, Shchur S, Favorov A, Boyko A, Favorova O. Pharmacogenetics of glatiramer acetate therapy for multiple sclerosis: the impact of genome-wide association studies identified disease risk loci. Pharmacogenomics 2017; 18:1563-1574. [PMID: 29095108 DOI: 10.2217/pgs-2017-0058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM Association analysis of genome-wide association studies (GWAS) identified multiple sclerosis (MS) risk genetic variants with glatiramer acetate (GA) treatment efficacy. PATIENTS & METHODS SNPs in 17 GWAS-identified immune response loci were analyzed in 296 Russian MS patients as possible markers of optimal GA treatment response for at least 2 years. RESULTS Alleles/genotypes of EOMES, CLEC16A, IL22RA2, PVT1 and HLA-DRB1 were associated by themselves with event-free phenotype during GA treatment for at least 2 years (p f = 0.032 - 0.00092). The biallelic combinations including EOMES, CLEC16A, IL22RA2, PVT1, TYK2, CD6, IL7RA and IRF8 genes were associated with response to GA with increased significance level (p f = 0.0060 - 1.1 × 10-5). The epistasic interactions or additive effects were observed between the components of the identified biallelic combinations. CONCLUSION We pinpointed the involvement of several GWAS-identified MS risk loci in GA therapy efficacy. These findings may be aggregated to predict the optimal GA response in MS patients.
Collapse
Affiliation(s)
- Olga Kulakova
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Vitalina Bashinskaya
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Ivan Kiselev
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Natalia Baulina
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Ekaterina Tsareva
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Ruslan Nikolaev
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Maxim Kozin
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Sergey Shchur
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Alexander Favorov
- Oncology Biostatistics & Bioinformatics, John Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Alexey Boyko
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Olga Favorova
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| |
Collapse
|
24
|
Graves JS, Barcellos LF, Simpson S, Belman A, Lin R, Taylor BV, Ponsonby AL, Dwyer T, Krupp L, Waubant E, van der Mei IAF. The multiple sclerosis risk allele within the AHI1 gene is associated with relapses in children and adults. Mult Scler Relat Disord 2017; 19:161-165. [PMID: 29409597 DOI: 10.1016/j.msard.2017.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/12/2017] [Accepted: 10/09/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND While common variant non-HLA (human leukocyte antigen) alleles have been associated with MS risk, their role in disease course is less clear. We sought to determine whether established multiple sclerosis (MS) genetic susceptibility factors are associated with relapse rate in children and an independent cohort of adults with MS. METHODS Genotyping was performed for 182 children with MS or clinically isolated syndrome with high risk for MS from two Pediatric MS Centers. They were prospectively followed for relapses. Fifty-two non-HLA MS susceptibility single nucleotide polymorphisms (SNPs) were evaluated for association with relapse rate. Cox regression models were adjusted for sex, genetic ancestry, disease-modifying therapy (DMT), 25-OH vitamin D level and HLA-DRB1*15:01/03 status. Investigation of pediatric subject SNP results was performed using a second cohort of 141 adult MS subjects of Northern European ancestry from the Southern Tasmanian Multiple Sclerosis Longitudinal Study. RESULTS For pediatric subjects, 408 relapses were captured over 622 patient-years of follow-up. Four non-HLA risk SNPs (rs11154801, rs650258, rs12212193, rs2303759) were associated with relapses (p < 0.01) in the pediatric subjects. After adjustment for genetic ancestry, sex, age, vitamin D level, DMT use and HLA-DRB1*15 status, having two copies of the MS risk allele within AHI1 (rs11154801) was associated with increased relapses among children (HR = 1.75,95%CI = 1.18-2.48, p = 0.006) and this result was also observed among adults (HR = 1.81,95%CI = 1.05-3.03, p = 0.026). CONCLUSIONS Our results suggest that the MS genetic risk variant within the gene AHI1 may contribute to disease course in addition to disease susceptibility.
Collapse
Affiliation(s)
- Jennifer S Graves
- UCSF Pediatric MS Center, San Francisco, CA, USA; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Lisa F Barcellos
- Genetic Epidemiology and Genomics Lab, School of Public Health, and California Institute of Quantitative Biosciences, UC Berkeley, Berkeley, CA, USA; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Steve Simpson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Anita Belman
- National Pediatric MS Center, Stonybrook, NY, USA; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Rui Lin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia; Guangxi Center for Disease Prevention and Control, Nanning, China; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Bruce V Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Anne-Louise Ponsonby
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Terence Dwyer
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Lauren Krupp
- National Pediatric MS Center, Stonybrook, NY, USA; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Emmanuelle Waubant
- UCSF Pediatric MS Center, San Francisco, CA, USA; School of Medicine, University of Tasmania, Hobart, Australia.
| | - Ingrid A F van der Mei
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia; School of Medicine, University of Tasmania, Hobart, Australia.
| |
Collapse
|
25
|
Ye MH, Bao H, Meng Y, Guan LL, Stothard P, Plastow G. Comparative transcriptomic analysis of porcine peripheral blood reveals differentially expressed genes from the cytokine-cytokine receptor interaction pathway related to health status. Genome 2017; 60:1021-1028. [PMID: 28763624 DOI: 10.1139/gen-2017-0074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While some research has looked into the host genetic response in pigs challenged with specific viruses or bacteria, few studies have explored the expression changes of transcripts in the peripheral blood of sick pigs that may be infected with multiple pathogens on farms. In this study, the architecture of the peripheral blood transcriptome of 64 Duroc sired commercial pigs, including 18 healthy animals at entry to a growing facility (set as a control) and 23 pairs of samples from healthy and sick pen mates, was generated using RNA-Seq technology. In total, 246 differentially expressed genes were identified to be specific to the sick animals. Functional enrichment analysis for those genes revealed that the over-represented gene ontology terms for the biological processes category were exclusively immune activity related. The cytokine-cytokine receptor interaction pathway was significantly enriched. Nine functional genes from this pathway encoding members (as well as their receptors) of the interleukins, chemokines, tumor necrosis factors, colony stimulating factors, activins, and interferons exhibited significant transcriptional alteration in sick animals. Our results suggest a subset of novel marker genes that may be useful candidate genes in the evaluation and prediction of health status in pigs under commercial production conditions.
Collapse
Affiliation(s)
- M H Ye
- a Department of Agricultural, Food and Nutritional Science, University of Alberta, 1400 College Plaza, 8215 112 Street, Edmonton, AB T6G 2C8, Canada.,b College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - H Bao
- a Department of Agricultural, Food and Nutritional Science, University of Alberta, 1400 College Plaza, 8215 112 Street, Edmonton, AB T6G 2C8, Canada
| | - Y Meng
- a Department of Agricultural, Food and Nutritional Science, University of Alberta, 1400 College Plaza, 8215 112 Street, Edmonton, AB T6G 2C8, Canada
| | - L L Guan
- a Department of Agricultural, Food and Nutritional Science, University of Alberta, 1400 College Plaza, 8215 112 Street, Edmonton, AB T6G 2C8, Canada
| | - P Stothard
- a Department of Agricultural, Food and Nutritional Science, University of Alberta, 1400 College Plaza, 8215 112 Street, Edmonton, AB T6G 2C8, Canada
| | - G Plastow
- a Department of Agricultural, Food and Nutritional Science, University of Alberta, 1400 College Plaza, 8215 112 Street, Edmonton, AB T6G 2C8, Canada
| |
Collapse
|
26
|
Lindahl H, Martini E, Brauner S, Nikamo P, Gallais Serezal I, Guerreiro-Cacais AO, Jagodic M, Eidsmo L, Ståhle M, Olsson T. IL-22 binding protein regulates murine skin inflammation. Exp Dermatol 2017; 26:444-446. [DOI: 10.1111/exd.13225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2016] [Indexed: 01/21/2023]
Affiliation(s)
- Hannes Lindahl
- Neuroimmunology Unit; Department of Clinical Neuroscience; Center for Molecular Medicine; Karolinska Institutet; Stockholm Sweden
| | - Elisa Martini
- Unit of Dermatology and Venereology; Department of Medicine; Karolinska lnstitutet, Karolinska University Hospital; Stockholm Sweden
| | - Susanna Brauner
- Neuroimmunology Unit; Department of Clinical Neuroscience; Center for Molecular Medicine; Karolinska Institutet; Stockholm Sweden
| | - Pernilla Nikamo
- Unit of Dermatology and Venereology; Department of Medicine; Karolinska lnstitutet, Karolinska University Hospital; Stockholm Sweden
| | - Irène Gallais Serezal
- Unit of Dermatology and Venereology; Department of Medicine; Karolinska lnstitutet, Karolinska University Hospital; Stockholm Sweden
| | - Andre Ortlieb Guerreiro-Cacais
- Neuroimmunology Unit; Department of Clinical Neuroscience; Center for Molecular Medicine; Karolinska Institutet; Stockholm Sweden
| | - Maja Jagodic
- Neuroimmunology Unit; Department of Clinical Neuroscience; Center for Molecular Medicine; Karolinska Institutet; Stockholm Sweden
| | - Liv Eidsmo
- Unit of Dermatology and Venereology; Department of Medicine; Karolinska lnstitutet, Karolinska University Hospital; Stockholm Sweden
| | - Mona Ståhle
- Unit of Dermatology and Venereology; Department of Medicine; Karolinska lnstitutet, Karolinska University Hospital; Stockholm Sweden
| | - Tomas Olsson
- Neuroimmunology Unit; Department of Clinical Neuroscience; Center for Molecular Medicine; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
27
|
Abstract
The cytokine interleukin-22 (IL-22), which is a member of the IL-10 family, is produced exclusively by immune cells and activates signal transducer and activator of transcription 3 (STAT3) in nonimmune cells, such as hepatocytes, keratinocytes, and colonic epithelial cells, to drive various processes central to tissue homeostasis and immunosurveillance. Dysregulation of IL-22 signaling causes inflammatory diseases. IL-22 binding protein (IL-22BP; encoded by IL22RA2) is a soluble IL-22 receptor, which antagonizes IL-22 activity and has genetic associations with autoimmune diseases. Humans have three IL-22BP isoforms, IL-22BPi1 to IL-22BPi3, which are generated by alternative splicing; mice only have an IL-22BPi2 homolog. We showed that, although IL-22BPi3 had less inhibitory activity than IL-22BPi2, IL-22BPi3 was more abundant in various human tissues under homeostatic conditions. IL-22BPi2 was more effective than IL-22BPi3 at blocking the contribution of IL-22 to cooperative gene induction with the inflammatory cytokine IL-17, which is often present with IL-22 in autoimmune settings. In addition, we found that IL-22BPi1 was not secreted and therefore failed to antagonize IL-22 signaling. Furthermore, IL-22BPi2 was the only isoform that was increased in abundance when myeloid cells were activated by Toll-like receptor 2 signaling or retinoic acid, a maturation factor for myeloid cells. These data suggest that the human IL-22BP isoforms have distinct spatial and temporal roles and coordinately fine-tune IL-22-dependent STAT3 responses in tissues as a type of rheostat.
Collapse
Affiliation(s)
- Chrissie Lim
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - MeeAe Hong
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Ram Savan
- Department of Immunology, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
28
|
Perusina Lanfranca M, Lin Y, Fang J, Zou W, Frankel T. Biological and pathological activities of interleukin-22. J Mol Med (Berl) 2016; 94:523-34. [PMID: 26923718 PMCID: PMC4860114 DOI: 10.1007/s00109-016-1391-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/17/2015] [Accepted: 01/21/2016] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-22, a member of the IL-10 family, is a cytokine secreted by several types of immune cells including IL-22(+)CD4(+) T cells (Th22) and IL-22 expressing innate leukocytes (ILC22). Recent studies have demonstrated that IL-22 is a key component in mucosal barrier defense, tissue repair, epithelial cell survival, and proliferation. Furthermore, accumulating evidence has defined both protective and pathogenic properties of IL-22 in a number of conditions including autoimmune disease, infection, and malignancy. In this review, we summarize the expression and signaling pathway and functional characteristics of the IL-22 and IL-22 receptor axis in physiological and pathological scenarios and discuss the potential to target IL-22 signaling to treat human diseases.
Collapse
Affiliation(s)
- Mirna Perusina Lanfranca
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Yanwei Lin
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, 200001, China
| | - Jingyuan Fang
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, 200001, China
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
- The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
- Graduate Programs in Immunology and Tumor Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Timothy Frankel
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
- The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
29
|
Guerreiro-Cacais AO, Laaksonen H, Flytzani S, N'diaye M, Olsson T, Jagodic M. Translational utility of experimental autoimmune encephalomyelitis: recent developments. J Inflamm Res 2015; 8:211-25. [PMID: 26622189 PMCID: PMC4654535 DOI: 10.2147/jir.s76707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Multiple sclerosis (MS) is a complex autoimmune condition with firmly established genetic and environmental components. Genome-wide association studies (GWAS) have revealed a large number of genetic polymorphisms in the vicinity of, and within, genes that associate to disease. However, the significance of these single-nucleotide polymorphisms in disease and possible mechanisms of action remain, with a few exceptions, to be established. While the animal model for MS, experimental autoimmune encephalomyelitis (EAE), has been instrumental in understanding immunity in general and mechanisms of MS disease in particular, much of the translational information gathered from the model in terms of treatment development (glatiramer acetate and natalizumab) has been extensively summarized. In this review, we would thus like to cover the work done in EAE from a GWAS perspective, highlighting the research that has addressed the role of different GWAS genes and their pathways in EAE pathogenesis. Understanding the contribution of these pathways to disease might allow for the stratification of disease subphenotypes in patients and in turn open the possibility for new and individualized treatment approaches in the future.
Collapse
Affiliation(s)
- Andre Ortlieb Guerreiro-Cacais
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Laaksonen
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sevasti Flytzani
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marie N'diaye
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maja Jagodic
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Perriard G, Mathias A, Enz L, Canales M, Schluep M, Gentner M, Schaeren-Wiemers N, Du Pasquier RA. Interleukin-22 is increased in multiple sclerosis patients and targets astrocytes. J Neuroinflammation 2015; 12:119. [PMID: 26077779 PMCID: PMC4480507 DOI: 10.1186/s12974-015-0335-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/03/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Increasing evidences link T helper 17 (Th17) cells with multiple sclerosis (MS). In this context, interleukin-22 (IL-22), a Th17-linked cytokine, has been implicated in blood brain barrier breakdown and lymphocyte infiltration. Furthermore, polymorphism between MS patients and controls has been recently described in the gene coding for IL-22 binding protein (IL-22BP). Here, we aimed to better characterize IL-22 in the context of MS. METHODS IL-22 and IL-22BP expressions were assessed by ELISA and qPCR in the following compartments of MS patients and control subjects: (1) the serum, (2) the cerebrospinal fluid, and (3) immune cells of peripheral blood. Identification of the IL-22 receptor subunit, IL-22R1, was performed by immunohistochemistry and immunofluorescence in human brain tissues and human primary astrocytes. The role of IL-22 on human primary astrocytes was evaluated using 7-AAD and annexin V, markers of cell viability and apoptosis, respectively. RESULTS In a cohort of 141 MS patients and healthy control (HC) subjects, we found that serum levels of IL-22 were significantly higher in relapsing MS patients than in HC but also remitting and progressive MS patients. Monocytes and monocyte-derived dendritic cells contained an enhanced expression of mRNA coding for IL-22BP as compared to HC. Using immunohistochemistry and confocal microscopy, we found that IL-22 and its receptor were detected on astrocytes of brain tissues from both control subjects and MS patients, although in the latter, the expression was higher around blood vessels and in MS plaques. Cytometry-based functional assays revealed that addition of IL-22 improved the survival of human primary astrocytes. Furthermore, tumor necrosis factor α-treated astrocytes had a better long-term survival capacity upon IL-22 co-treatment. This protective effect of IL-22 seemed to be conferred, at least partially, by a decreased apoptosis. CONCLUSIONS We show that (1) there is a dysregulation in the expression of IL-22 and its antagonist, IL-22BP, in MS patients, (2) IL-22 targets specifically astrocytes in the human brain, and (3) this cytokine confers an increased survival of the latter cells.
Collapse
Affiliation(s)
- Guillaume Perriard
- Laboratory of Neuroimmunology, Center of Research in Neurosciences, Department of Clinical Neurosciences and Service of Immunology and Allergy, Department of Medicine, CHUV, 1011, Lausanne, Switzerland
| | - Amandine Mathias
- Laboratory of Neuroimmunology, Center of Research in Neurosciences, Department of Clinical Neurosciences and Service of Immunology and Allergy, Department of Medicine, CHUV, 1011, Lausanne, Switzerland
| | - Lukas Enz
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Mathieu Canales
- Laboratory of Neuroimmunology, Center of Research in Neurosciences, Department of Clinical Neurosciences and Service of Immunology and Allergy, Department of Medicine, CHUV, 1011, Lausanne, Switzerland
| | - Myriam Schluep
- Service of Neurology, Department of Clinical Neurosciences, CHUV BH-10/131, 46, rue du Bugnon, 1011, Lausanne, Switzerland
| | - Melanie Gentner
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Nicole Schaeren-Wiemers
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Renaud A Du Pasquier
- Laboratory of Neuroimmunology, Center of Research in Neurosciences, Department of Clinical Neurosciences and Service of Immunology and Allergy, Department of Medicine, CHUV, 1011, Lausanne, Switzerland.
- Service of Neurology, Department of Clinical Neurosciences, CHUV BH-10/131, 46, rue du Bugnon, 1011, Lausanne, Switzerland.
| |
Collapse
|
31
|
Didonna A, Oksenberg JR. Genetic determinants of risk and progression in multiple sclerosis. Clin Chim Acta 2015; 449:16-22. [PMID: 25661088 DOI: 10.1016/j.cca.2015.01.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/21/2015] [Indexed: 01/07/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disease that represents a primary cause of neurological disability in the young adult population. Converging evidence supports the importance of genetic determinants for MS etiology. However, with the exception of the major histocompatibility complex, their nature has been elusive for more than 20 years. In the last decade, the advent of large genome-wide association studies has significantly improved our understanding of the disease, leading to the golden era of MS genetic research. To date more than 110 genetic variants have been firmly associated to an increased risk of developing MS. A large part of these variants tag genes involved in the regulation of immune response and several of them are shared with other autoimmune diseases, suggesting a common etiological root for this class of disorders. Despite the impressive body of data obtained in the last years, we are still far from fully decoding MS genetic complexity. For example, we ignore how these genetic factors interact with each other and with the environment. Thus, the biggest challenge for the next era of MS research will consist in identifying and characterizing the molecular mechanisms and the cellular pathways in which these risk variants play a role.
Collapse
Affiliation(s)
- Alessandro Didonna
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jorge R Oksenberg
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|