1
|
Yang F, Li X, Wang J, Duan Z, Ren C, Guo P, Kong Y, Bi M, Zhang Y. Identification of lipid metabolism-related gene markers and construction of a diagnostic model for multiple sclerosis: An integrated analysis by bioinformatics and machine learning. Anal Biochem 2025; 700:115781. [PMID: 39855613 DOI: 10.1016/j.ab.2025.115781] [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: 09/13/2024] [Revised: 12/20/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune inflammatory disorder that causes neurological disability. Dysregulated lipid metabolism contributes to the pathogenesis of MS. This study aimed to identify lipid metabolism-related gene markers and construct a diagnostic model for MS. METHODS Gene expression profiles for MS were obtained from the Gene Expression Omnibus database. Differentially expressed lipid metabolism-related genes (LMRGs) were identified and performed functional enrichment analysis. Least absolute shrinkage and selection operator (LASSO), random forest (RF), and protein-protein interaction (PPI) analysis were employed to screen hub genes. The predictive power of hub genes was evaluated using receiver operating characteristic (ROC) curves. We developed an artificial neural network (ANN) model and validated its performance in three test sets. Immune cell infiltration analysis, Gene set enrichment analysis, and ceRNA network construction were performed to explore the role of lipid metabolism in the pathogenesis of MS. Drugs prediction and molecular docking were utilized to identify potential therapeutic drugs. RESULTS We identified 40 differentially expressed LMRGs, with significant enrichment in Arachidonic acid metabolism, Steroid hormone biosynthesis, Fatty acid elongation, and Sphingolipid metabolism. AKR1C3, NFKB1, and ABCA1 were identified as gene markers for MS, and their expression was upregulated in the MS group. The areas under the ROC curve (AUCs) for AKR1C3, NFKB1, and ABCA1 in the training set were 0.779, 0.703, and 0.726, respectively. The ANN model exhibited good discriminative ability in both the training and test sets, achieving an AUC of 0.826 on the training set and AUC values of 0.822, 0.890, and 0.833 on the test sets. Gamma.delta.T.cell, Natural.killer.T.cell, Plasmacytoid.dendritic.cell, Regulatory.T.cell, and Type.1.T.helper.cell were highly expressed in the MS group. A ceRNA network showed a complex regulatory interplay involving hub genes. Luteolin, isoflavone, and thalidomide had good binding affinities to the hub genes. CONCLUSION Our study emphasized the crucial role of lipid metabolism in MS, identifing AKR1C3, NFKB1, and ABCA1 as gene markers. The ANN model exhibited good performance on both the training and testing sets. These findings offer valuable insights into the molecular mechanisms underlying MS, and establish a scientific foundation for future research.
Collapse
Affiliation(s)
- Fangjie Yang
- Rehabilitation Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xinmin Li
- School of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jing Wang
- Rehabilitation Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zhenfei Duan
- Rehabilitation Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Chunlin Ren
- Rehabilitation Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Pengxue Guo
- Rehabilitation Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yuting Kong
- Rehabilitation Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Mengyao Bi
- Rehabilitation Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yasu Zhang
- Rehabilitation Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| |
Collapse
|
2
|
Nabeel Mustafa A, Salih Mahdi M, Ballal S, Chahar M, Verma R, Ali Al-Nuaimi AM, Kumar MR, Kadhim A Al-Hussein R, Adil M, Jasem Jawad M. Netrin-1: Key insights in neural development and disorders. Tissue Cell 2025; 93:102678. [PMID: 39719818 DOI: 10.1016/j.tice.2024.102678] [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: 08/27/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/26/2024]
Abstract
Netrin-1, an essential extracellular protein, has gained significant attention due to its pivotal role in guiding axon and cell migration during embryonic development. The fundamental significance of netrin-1 in developmental biology is reflected in its high conservation across different species as a part of the netrin family. The bifunctional nature of netrin-1 demonstrates its functional versatility, as it can function as either a repellent or an attractant according to the context and the expressed receptors on the target cells including the deleted in colorectal cancer (DCC), the uncoordinated-5 (UNC5), DSCAM, Neogenin-1, Adenosine A2b and Draxin receptors. By directing axonal growth cones toward the appropriate targets, netrin-1 is a critical actor in the formation of the intricate architecture of the nervous system. Netrin-1 is believed to be involved in additional biological and pathological processes in addition to its traditional function in neural development. The behavior of a diverse array of cell types is influenced by controlling cell adhesion and movement, which is impacted by netrin-1. It is a molecule of interest in both developmental biology and clinical research because of its involvement in angiogenesis, tumorigenesis, inflammation, and tissue regeneration, as confirmed by recent studies. The therapeutic capability of netrin-1 in disorders such as cancer, neurodegenerative disorders, and cardiovascular diseases warrants significant attention.
Collapse
Affiliation(s)
| | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bengaluru, Karnataka, India
| | - Mamata Chahar
- Department of Chemistry, NIMS University, Jaipur, Rajasthan, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | | | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | | | | | | |
Collapse
|
3
|
Patalano SD, Fuxman Bass P, Fuxman Bass JI. Transcription factors in the development and treatment of immune disorders. Transcription 2025; 16:118-140. [PMID: 38100543 PMCID: PMC11970766 DOI: 10.1080/21541264.2023.2294623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Immune function is highly controlled at the transcriptional level by the binding of transcription factors (TFs) to promoter and enhancer elements. Several TF families play major roles in immune gene expression, including NF-κB, STAT, IRF, AP-1, NRs, and NFAT, which trigger anti-pathogen responses, promote cell differentiation, and maintain immune system homeostasis. Aberrant expression, activation, or sequence of isoforms and variants of these TFs can result in autoimmune and inflammatory diseases as well as hematological and solid tumor cancers. For this reason, TFs have become attractive drug targets, even though most were previously deemed "undruggable" due to their lack of small molecule binding pockets and the presence of intrinsically disordered regions. However, several aspects of TF structure and function can be targeted for therapeutic intervention, such as ligand-binding domains, protein-protein interactions between TFs and with cofactors, TF-DNA binding, TF stability, upstream signaling pathways, and TF expression. In this review, we provide an overview of each of the important TF families, how they function in immunity, and some related diseases they are involved in. Additionally, we discuss the ways of targeting TFs with drugs along with recent research developments in these areas and their clinical applications, followed by the advantages and disadvantages of targeting TFs for the treatment of immune disorders.
Collapse
Affiliation(s)
- Samantha D. Patalano
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Paula Fuxman Bass
- Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I. Fuxman Bass
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
4
|
Justus CR, Marie MA, Sanderlin EJ, Yang LV. The Roles of Proton-Sensing G-Protein-Coupled Receptors in Inflammation and Cancer. Genes (Basel) 2024; 15:1151. [PMID: 39336742 PMCID: PMC11431078 DOI: 10.3390/genes15091151] [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: 08/01/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The precise regulation of pH homeostasis is crucial for normal physiology. However, in tissue microenvironments, it can be impacted by pathological conditions such as inflammation and cancer. Due to the overproduction and accumulation of acids (protons), the extracellular pH is characteristically more acidic in inflamed tissues and tumors in comparison to normal tissues. A family of proton-sensing G-protein-coupled receptors (GPCRs) has been identified as molecular sensors for cells responding to acidic tissue microenvironments. Herein, we review the current research progress pertaining to these proton-sensing GPCRs, including GPR4, GPR65 (TDAG8), and GPR68 (OGR1), in inflammation and cancer. Growing evidence suggests that GPR4 and GPR68 are mainly pro-inflammatory, whereas GPR65 is primarily anti-inflammatory, in various inflammatory disorders. Both anti- and pro-tumorigenic effects have been reported for this family of receptors. Moreover, antagonists and agonists targeting proton-sensing GPCRs have been developed and evaluated in preclinical models. Further research is warranted to better understand the roles of these proton-sensing GPCRs in pathophysiology and is required in order to exploit them as potential therapeutic targets for disease treatment.
Collapse
Affiliation(s)
- Calvin R Justus
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Mona A Marie
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Edward J Sanderlin
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Li V Yang
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
5
|
Tian H, Huang D, Wang J, Li H, Gao J, Zhong Y, Xia L, Zhang A, Lin Z, Ke X. The role of the "gut microbiota-mitochondria" crosstalk in the pathogenesis of multiple sclerosis. Front Microbiol 2024; 15:1404995. [PMID: 38741740 PMCID: PMC11089144 DOI: 10.3389/fmicb.2024.1404995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024] Open
Abstract
Multiple Sclerosis (MS) is a neurologic autoimmune disease whose exact pathophysiologic mechanisms remain to be elucidated. Recent studies have shown that the onset and progression of MS are associated with dysbiosis of the gut microbiota. Similarly, a large body of evidence suggests that mitochondrial dysfunction may also have a significant impact on the development of MS. Endosymbiotic theory has found that human mitochondria are microbial in origin and share similar biological characteristics with the gut microbiota. Therefore, gut microbiota and mitochondrial function crosstalk are relevant in the development of MS. However, the relationship between gut microbiota and mitochondrial function in the development of MS is not fully understood. Therefore, by synthesizing previous relevant literature, this paper focuses on the changes in gut microbiota and metabolite composition in the development of MS and the possible mechanisms of the crosstalk between gut microbiota and mitochondrial function in the progression of MS, to provide new therapeutic approaches for the prevention or reduction of MS based on this crosstalk.
Collapse
Affiliation(s)
- Huan Tian
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dunbing Huang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiaqi Wang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huaqiang Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiaxin Gao
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Zhong
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Libin Xia
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhonghua Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Rehabilitation Medicine Center, Fujian Provincial Hospital, Fuzhou, China
- Fujian Provincial Center for Geriatrics, Fujian Provincia Hospital, Fuzhou, China
| | - Xiaohua Ke
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
6
|
Hausmann M, Seuwen K, de Vallière C, Busch M, Ruiz PA, Rogler G. Role of pH-sensing receptors in colitis. Pflugers Arch 2024; 476:611-622. [PMID: 38514581 PMCID: PMC11006753 DOI: 10.1007/s00424-024-02943-y] [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: 12/26/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024]
Abstract
Low pH in the gut is associated with severe inflammation, fibrosis, and colorectal cancer (CRC) and is a hallmark of active inflammatory bowel disease (IBD). Subsequently, pH-sensing mechanisms are of interest for the understanding of IBD pathophysiology. Tissue hypoxia and acidosis-two contributing factors to disease pathophysiology-are linked to IBD, and understanding their interplay is highly relevant for the development of new therapeutic options. One member of the proton-sensing G protein-coupled receptor (GPCR) family, GPR65 (T-cell death-associated gene 8, TDAG8), was identified as a susceptibility gene for IBD in a large genome-wide association study. In response to acidic extracellular pH, GPR65 induces an anti-inflammatory response, whereas the two other proton-sensing receptors, GPR4 and GPR68 (ovarian cancer G protein-coupled receptor 1, OGR1), mediate pro-inflammatory responses. Here, we review the current knowledge on the role of these proton-sensing receptors in IBD and IBD-associated fibrosis and cancer, as well as colitis-associated cancer (CAC). We also describe emerging small molecule modulators of these receptors as therapeutic opportunities for the treatment of IBD.
Collapse
Affiliation(s)
- Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland.
| | - Klaus Seuwen
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Cheryl de Vallière
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Moana Busch
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Pedro A Ruiz
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| |
Collapse
|
7
|
Perren L, Busch M, Schuler C, Ruiz PA, Foti F, Weibel N, de Vallière C, Morsy Y, Seuwen K, Hausmann M, Rogler G. OGR1 (GPR68) and TDAG8 (GPR65) Have Antagonistic Effects in Models of Colonic Inflammation. Int J Mol Sci 2023; 24:14855. [PMID: 37834303 PMCID: PMC10573511 DOI: 10.3390/ijms241914855] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
G-protein-coupled receptors (GPRs), including pro-inflammatory ovarian cancer GPR1 (OGR1/GPR68) and anti-inflammatory T cell death-associated gene 8 (TDAG8/GPR65), are involved in pH sensing and linked to inflammatory bowel disease (IBD). OGR1 and TDAG8 show opposite effects. To determine which effect is predominant or physiologically more relevant, we deleted both receptors in models of intestinal inflammation. Combined Ogr1 and Tdag8 deficiency was assessed in spontaneous and acute murine colitis models. Disease severity was assessed using clinical scores. Colon samples were analyzed using quantitative polymerase chain reaction (qPCR) and flow cytometry (FACS). In acute colitis, Ogr1-deficient mice showed significantly decreased clinical scores compared with wildtype (WT) mice, while Tdag8-deficient mice and double knockout (KO) mice presented similar scores to WT. In Il-10-spontaneous colitis, Ogr1-deficient mice presented significantly decreased, and Tdag8-deficient mice had increased inflammation. In the Il10-/- × Ogr1-/- × Tdag8-/- triple KO mice, inflammation was significantly decreased compared with Tdag8-/-. Absence of Ogr1 reduced pro-inflammatory cytokines in Tdag8-deficient mice. Tdag8-/- had significantly more IFNγ+ T-lymphocytes and IL-23 T-helper cells in the colon compared with WT. The absence of OGR1 significantly alleviates the intestinal damage mediated by the lack of functional TDAG8. Both OGR1 and TDAG8 represent potential new targets for therapeutic intervention.
Collapse
|
8
|
Ortiz GG, Torres-Mendoza BMG, Ramírez-Jirano J, Marquez-Pedroza J, Hernández-Cruz JJ, Mireles-Ramirez MA, Torres-Sánchez ED. Genetic Basis of Inflammatory Demyelinating Diseases of the Central Nervous System: Multiple Sclerosis and Neuromyelitis Optica Spectrum. Genes (Basel) 2023; 14:1319. [PMID: 37510224 PMCID: PMC10379341 DOI: 10.3390/genes14071319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Demyelinating diseases alter myelin or the coating surrounding most nerve fibers in the central and peripheral nervous systems. The grouping of human central nervous system demyelinating disorders today includes multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD) as distinct disease categories. Each disease is caused by a complex combination of genetic and environmental variables, many involving an autoimmune response. Even though these conditions are fundamentally similar, research into genetic factors, their unique clinical manifestations, and lesion pathology has helped with differential diagnosis and disease pathogenesis knowledge. This review aims to synthesize the genetic approaches that explain the differential susceptibility between these diseases, explore the overlapping clinical features, and pathological findings, discuss existing and emerging hypotheses on the etiology of demyelination, and assess recent pathogenicity studies and their implications for human demyelination. This review presents critical information from previous studies on the disease, which asks several questions to understand the gaps in research in this field.
Collapse
Affiliation(s)
- Genaro Gabriel Ortiz
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in Medicine Hospital, Civil University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara 44329, Jalisco, Mexico
| | - Blanca M G Torres-Mendoza
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in Medicine Hospital, Civil University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Neurosciences Division, Western Biomedical Research Center, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Javier Ramírez-Jirano
- Neurosciences Division, Western Biomedical Research Center, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Jazmin Marquez-Pedroza
- Neurosciences Division, Western Biomedical Research Center, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Guadalajara 44340, Jalisco, Mexico
- Coordination of Academic Activities, Western Biomedical Research Center, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Guadalajara 44340, Jalisco, Mexico
| | - José J Hernández-Cruz
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara 44329, Jalisco, Mexico
| | - Mario A Mireles-Ramirez
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara 44329, Jalisco, Mexico
| | - Erandis D Torres-Sánchez
- Department of Medical and Life Sciences, University Center of la Cienega, University of Guadalajara, Ocotlan 47820, Jalisco, Mexico
| |
Collapse
|
9
|
da Silva EV, Fontes-Dantas FL, Dantas TV, Dutra A, Nascimento OJM, Alves-Leon SV. Shared Molecular Signatures Across Zika Virus Infection and Multiple Sclerosis Highlight AP-1 Transcription Factor as a Potential Player in Post-ZIKV MS-Like Phenotypes. Mol Neurobiol 2023:10.1007/s12035-023-03305-y. [PMID: 37046138 DOI: 10.1007/s12035-023-03305-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/08/2023] [Indexed: 04/14/2023]
Abstract
Zika virus (ZIKV) is an arbovirus of the Flaviviridae genus that has rapidly disseminated from across the Pacific to the Americas. Robust evidence has indicated a crucial role of ZIKV in congenital virus syndrome, including neonatal microcephaly. Moreover, emerging evidence suggests an association between ZIKV infection and the development of an extensive spectrum of central nervous system inflammatory demyelinating diseases (CNS IDD), such as multiple sclerosis-like clinical phenotypes. However, the underlying mechanisms of host-pathogen neuro-immune interactions remain to be elucidated. This study aimed to identify common transcriptional signatures between multiple sclerosis (MS) and ZIKV infection to generate molecular interaction networks, thereby leading to the identification of deregulated processes and pathways, which could give an insight of these underlying molecular mechanisms. Our investigation included publicly available transcriptomic data from MS patients in either relapse or remission (RR-MS) and datasets of subjects acutely infected by ZIKV for both immune peripheral cells and central nervous system cells. The protein-protein interaction (PPI) analysis showed upregulated AP-1 transcription factors (JUN and FOS) among the top hub and bottleneck genes in RR-MS and ZIKV data. Gene enrichment analysis retrieved a remarkable presence of ontologies and pathways linked to oxidative stress responses, immune cell function, inflammation, interleukin signaling, cell division, and transcriptional regulation commonly enriched in both scenarios. Considering the recent findings concerning AP-1 function in immunological tolerance breakdown, regulation of inflammation, and its function as an oxidative stress sensor, we postulate that the ZIKV trigger may contribute as a boost for the activation of such AP-1-regulated mechanisms that could favor the development of MS-like phenotypes following ZIKV infection in a genetically susceptible individual.
Collapse
Affiliation(s)
- Elielson Veloso da Silva
- Laboratório de Neurociências Translacional, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de pós-graduação em Medicina (Neurologia/Neurociências), Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Fabrícia Lima Fontes-Dantas
- Laboratório de Neurociências Translacional, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcântara Gomes, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thiago Viana Dantas
- Programa de Engenharia de Sistemas e Computação-COPPE, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amanda Dutra
- Laboratório de Neurociências Translacional, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Osvaldo J M Nascimento
- Programa de pós-graduação em Medicina (Neurologia/Neurociências), Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Soniza Vieira Alves-Leon
- Laboratório de Neurociências Translacional, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil.
- Hospital Universitário Clementino Fraga Filho, Centro de Referência em Doenças Inflamatórias Desmielinizantes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
10
|
Park SY, Kim MW, Kang JH, Jung HJ, Hwang JH, Yang SJ, Woo JK, Jeon Y, Lee H, Yoon YS, Seong JK, Oh SH. Novel NF-κB reporter mouse for the non-invasive monitoring of inflammatory diseases. Sci Rep 2023; 13:3556. [PMID: 36864088 PMCID: PMC9981691 DOI: 10.1038/s41598-023-29689-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/08/2023] [Indexed: 03/04/2023] Open
Abstract
Bioluminescence imaging is useful for non-invasively monitoring inflammatory reactions associated with disease progression, and since NF-κB is a pivotal transcription factor that alters expressions of inflammatory genes, we generated novel NF-κB luciferase reporter (NF-κB-Luc) mice to understand the dynamics of inflammatory responses in whole body, and also in various type of cells by crossing NF-κB-Luc mice with cell-type specific Cre expressing mice (NF-κB-Luc:[Cre]). Bioluminescence intensity was significantly increased in NF-κB-Luc (NKL) mice exposed to inflammatory stimuli (PMA or LPS). Crossing NF-κB-Luc mice with Alb-cre mice or Lyz-cre mice generated NF-κB-Luc:Alb (NKLA) and NF-κB-Luc:Lyz2 (NKLL) mice, respectively. NKLA and NKLL mice showed enhanced bioluminescence in liver and macrophages, respectively. To confirm that our reporter mice could be utilized for the non-invasive monitoring of inflammation in preclinical models, we conducted a DSS-induced colitis model and a CDAHFD-induced NASH model in our reporter mice. In both models, our reporter mice reflected the development of these diseases over time. In conclusion, we believe that our novel reporter mouse can be utilized as a non-invasive monitoring platform for inflammatory diseases.
Collapse
Affiliation(s)
- Se Yong Park
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min Woo Kim
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Ju-Hee Kang
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Hyun Jin Jung
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Jung Ho Hwang
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Soo Jung Yang
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Jong Kyu Woo
- Korea Mouse Phenotyping Center (KMPC), College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yoon Jeon
- Graduate School of Cancer Science and Policy, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Yeo Sung Yoon
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center (KMPC), College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| | - Seung Hyun Oh
- College of Pharmacy, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
11
|
Monos DS, Rajalingam R. The Major Histocompatibility Complex. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
12
|
Miyauchi E, Shimokawa C, Steimle A, Desai MS, Ohno H. The impact of the gut microbiome on extra-intestinal autoimmune diseases. Nat Rev Immunol 2023; 23:9-23. [PMID: 35534624 DOI: 10.1038/s41577-022-00727-y] [Citation(s) in RCA: 181] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2022] [Indexed: 02/08/2023]
Abstract
The prevalence of autoimmune diseases (ADs) worldwide has rapidly increased over the past few decades. Thus, in addition to the classical risk factors for ADs, such as genetic polymorphisms, infections and smoking, environmental triggers have been considered. Recent sequencing-based approaches have revealed that patients with extra-intestinal ADs, such as multiple sclerosis, rheumatoid arthritis, type 1 diabetes and systemic lupus erythematosus, have distinct gut microbiota compositions compared to healthy controls. Faecal microbiota transplantation or inoculation with specific microbes in animal models of ADs support the hypothesis that alterations of gut microbiota influence autoimmune responses and disease outcome. Here, we describe the compositional and functional changes in the gut microbiota in patients with extra-intestinal AD and discuss how the gut microbiota affects immunity. Moreover, we examine how the gut microbiota might be modulated in patients with ADs as a potential preventive or therapeutic approach.
Collapse
Affiliation(s)
- Eiji Miyauchi
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Institute for Molecular and Cellular Regulation, Gunma University, Haebashi, Gunma, Japan
| | - Chikako Shimokawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Parasitology, National Institute of Infectious Disease, Tokyo, Japan
| | - Alex Steimle
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| | - Hiroshi Ohno
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan.
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa, Japan.
- Laboratory for Immune Regulation, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan.
| |
Collapse
|
13
|
dos Santos de Souza RBM, Soares NMM, Bastos TS, Kaelle GCB, de Oliveira SG, Félix FAP. Effects of dietary supplementation with a blend of functional oils to fecal microbiota, and inflammatory and oxidative responses, of dogs submitted to a periodontal surgical challenge. Anim Feed Sci Technol 2022. [DOI: 10.1016/j.anifeedsci.2022.115527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Li T, Bou-Dargham MJ, Fultang N, Li X, Pear WS, Sun H, Chen YH. c-Rel-dependent monocytes are potent immune suppressor cells in cancer. J Leukoc Biol 2022; 112:845-859. [PMID: 35694784 PMCID: PMC9530019 DOI: 10.1002/jlb.1ma0422-518rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 04/25/2022] [Indexed: 11/08/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of leukocytes that are important for tumorigenesis and tumor immunotherapy. They comprise up to 10% of leukocytes in the blood of tumor patients and their depletion may be required for successful tumor immunotherapy. However, the identity of MDSCs remains obscure, primarily due to their heterogeneity and lack of a known lineage-specific transcription factor specifying their differentiation. Using single-cell transcriptomics and gene knockout approaches, we now describe a subset of murine and human myeloid suppressor cells, named rel-dependent monocytes (rMos), which are programmed by the transcription factor c-Rel of the NF-κB family. Unlike MDSCs described previously, the c-Rel-dependent monocytes expressed a high amount of the proinflammatory cytokine IL-1β together with a low level of suppressive molecule arginase 1. Both in vitro and in tumor-bearing mice, these c-Rel+ IL-1βhi Arg1- monocytes promoted tumor growth by potently suppressing T cell function and showed a strong migratory phenotype, all of which were impaired by c-Rel deficiency or inhibition. Mechanistic studies revealed that c-Rel controlled the expression of monocyte signature genes through a unique transcriptional complex called the c-Rel enhanceosome, and IL-1β-CCL2 crosstalk between tumor cells and the rel-dependent monocytes maintained the suppressive tumor microenvironment. Thus, c-Rel specifies the development of a suppressive monocyte population and could be selectively targeted for treating cancer.
Collapse
Affiliation(s)
- Ting Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mayassa J Bou-Dargham
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Norman Fultang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xinyuan Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Warren S Pear
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Honghong Sun
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Faculty of Pharmaceutical Sciences, CAS Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong, China
| |
Collapse
|
15
|
Roberti A, Chaffey LE, Greaves DR. NF-κB Signaling and Inflammation-Drug Repurposing to Treat Inflammatory Disorders? BIOLOGY 2022; 11:372. [PMID: 35336746 PMCID: PMC8945680 DOI: 10.3390/biology11030372] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022]
Abstract
NF-κB is a central mediator of inflammation, response to DNA damage and oxidative stress. As a result of its central role in so many important cellular processes, NF-κB dysregulation has been implicated in the pathology of important human diseases. NF-κB activation causes inappropriate inflammatory responses in diseases including rheumatoid arthritis (RA) and multiple sclerosis (MS). Thus, modulation of NF-κB signaling is being widely investigated as an approach to treat chronic inflammatory diseases, autoimmunity and cancer. The emergence of COVID-19 in late 2019, the subsequent pandemic and the huge clinical burden of patients with life-threatening SARS-CoV-2 pneumonia led to a massive scramble to repurpose existing medicines to treat lung inflammation in a wide range of healthcare systems. These efforts continue and have proven to be controversial. Drug repurposing strategies are a promising alternative to de novo drug development, as they minimize drug development timelines and reduce the risk of failure due to unexpected side effects. Different experimental approaches have been applied to identify existing medicines which inhibit NF-κB that could be repurposed as anti-inflammatory drugs.
Collapse
Affiliation(s)
| | | | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK; (A.R.); (L.E.C.)
| |
Collapse
|
16
|
Sato F, Nakamura Y, Katsuki A, Khadka S, Ahmad I, Omura S, Martinez NE, Tsunoda I. Curdlan, a Microbial β-Glucan, Has Contrasting Effects on Autoimmune and Viral Models of Multiple Sclerosis. Front Cell Infect Microbiol 2022; 12:805302. [PMID: 35198458 PMCID: PMC8859099 DOI: 10.3389/fcimb.2022.805302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disease characterized by inflammatory demyelination and axonal degeneration in the central nervous system (CNS). Bacterial and fungal infections have been associated with the development of MS; microbial components that are present in several microbes could contribute to MS pathogenesis. Among such components, curdlan is a microbial 1,3-β-glucan that can stimulate dendritic cells, and enhances T helper (Th) 17 responses. We determined whether curdlan administration could affect two animal models for MS: an autoimmune model, experimental autoimmune encephalomyelitis (EAE), and a viral model, Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD). We induced relapsing-remitting EAE by sensitizing SJL/J mice with the myelin proteolipid protein (PLP)139-151 peptide and found that curdlan treatment prior to PLP sensitization converted the clinical course of EAE into hyperacute EAE, in which the mice developed a progressive motor paralysis and died within 2 weeks. Curdlan-treated EAE mice had massive infiltration of T cells and neutrophils in the CNS with higher levels of Th17 and Th1 responses, compared with the control EAE mice. On the other hand, in TMEV-IDD, we found that curdlan treatment reduced the clinical scores and axonal degeneration without changes in inflammation or viral persistence in the CNS. In summary, although curdlan administration exacerbated the autoimmune MS model by enhancing inflammatory demyelination, it suppressed the viral MS model with reduced axonal degeneration. Therefore, microbial infections may play contrasting roles in MS depending on its etiology: autoimmunity versus viral infection.
Collapse
Affiliation(s)
- Fumitaka Sato
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health-Shreveport, Shreveport, LA, United States
| | - Yumina Nakamura
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Aoshi Katsuki
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Sundar Khadka
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Ijaz Ahmad
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Seiichi Omura
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health-Shreveport, Shreveport, LA, United States
| | - Nicholas E. Martinez
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health-Shreveport, Shreveport, LA, United States
| | - Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health-Shreveport, Shreveport, LA, United States
| |
Collapse
|
17
|
GPR65 (TDAG8) inhibits intestinal inflammation and colitis-associated colorectal cancer development in experimental mouse models. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166288. [PMID: 34628032 PMCID: PMC8629932 DOI: 10.1016/j.bbadis.2021.166288] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023]
Abstract
GPR65 (TDAG8) is a proton-sensing G protein-coupled receptor predominantly expressed in immune cells. Genome-wide association studies (GWAS) have identified GPR65 gene polymorphisms as an emerging risk factor for the development of inflammatory bowel disease (IBD). Patients with IBD have an elevated risk of developing colorectal cancer when compared to the general population. To study the role of GPR65 in intestinal inflammation and colitis-associated colorectal cancer (CAC), colitis and CAC were induced in GPR65 knockout (KO) and wild-type (WT) mice using dextran sulfate sodium (DSS) and azoxymethane (AOM)/DSS, respectively. Disease severity parameters such as fecal score, colon shortening, histopathology, and mesenteric lymph node enlargement were aggravated in GPR65 KO mice compared to WT mice treated with DSS. Elevated leukocyte infiltration and fibrosis were observed in the inflamed colon of GPR65 KO when compared to WT mice which may represent a cellular mechanism for the observed exacerbation of intestinal inflammation. In line with high expression of GPR65 in infiltrated leukocytes, GPR65 gene expression was increased in inflamed intestinal tissue samples of IBD patients compared to normal intestinal tissues. Moreover, colitis-associated colorectal cancer development was higher in GPR65 KO mice than WT mice when treated with AOM/DSS. Altogether, our data demonstrate that GPR65 suppresses intestinal inflammation and colitis-associated tumor development in murine colitis and CAC models, suggesting potentiation of GPR65 with agonists may have an anti-inflammatory therapeutic effect in IBD and reduce the risk of developing colitis-associated colorectal cancer.
Collapse
|
18
|
Bernardo-Faura M, Rinas M, Wirbel J, Pertsovskaya I, Pliaka V, Messinis DE, Vila G, Sakellaropoulos T, Faigle W, Stridh P, Behrens JR, Olsson T, Martin R, Paul F, Alexopoulos LG, Villoslada P, Saez-Rodriguez J. Prediction of combination therapies based on topological modeling of the immune signaling network in multiple sclerosis. Genome Med 2021; 13:117. [PMID: 34271980 PMCID: PMC8284018 DOI: 10.1186/s13073-021-00925-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/14/2021] [Indexed: 11/21/2022] Open
Abstract
Background Multiple sclerosis (MS) is a major health problem, leading to a significant disability and patient suffering. Although chronic activation of the immune system is a hallmark of the disease, its pathogenesis is poorly understood, while current treatments only ameliorate the disease and may produce severe side effects. Methods Here, we applied a network-based modeling approach based on phosphoproteomic data to uncover the differential activation in signaling wiring between healthy donors, untreated patients, and those under different treatments. Based in the patient-specific networks, we aimed to create a new approach to identify drug combinations that revert signaling to a healthy-like state. We performed ex vivo multiplexed phosphoproteomic assays upon perturbations with multiple drugs and ligands in primary immune cells from 169 subjects (MS patients, n=129 and matched healthy controls, n=40). Patients were either untreated or treated with fingolimod, natalizumab, interferon-β, glatiramer acetate, or the experimental therapy epigallocatechin gallate (EGCG). We generated for each donor a dynamic logic model by fitting a bespoke literature-derived network of MS-related pathways to the perturbation data. Last, we developed an approach based on network topology to identify deregulated interactions whose activity could be reverted to a “healthy-like” status by combination therapy. The experimental autoimmune encephalomyelitis (EAE) mouse model of MS was used to validate the prediction of combination therapies. Results Analysis of the models uncovered features of healthy-, disease-, and drug-specific signaling networks. We predicted several combinations with approved MS drugs that could revert signaling to a healthy-like state. Specifically, TGF-β activated kinase 1 (TAK1) kinase, involved in Transforming growth factor β-1 proprotein (TGF-β), Toll-like receptor, B cell receptor, and response to inflammation pathways, was found to be highly deregulated and co-druggable with all MS drugs studied. One of these predicted combinations, fingolimod with a TAK1 inhibitor, was validated in an animal model of MS. Conclusions Our approach based on donor-specific signaling networks enables prediction of targets for combination therapy for MS and other complex diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-021-00925-8.
Collapse
Affiliation(s)
- Marti Bernardo-Faura
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK.,Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Barcelona, Spain
| | - Melanie Rinas
- Joint Research Center for Computational Biomedicine (JRC-COMBINE), Faculty of Medicine, RWTH-Aachen University, Aachen, Germany
| | - Jakob Wirbel
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK.,Joint Research Center for Computational Biomedicine (JRC-COMBINE), Faculty of Medicine, RWTH-Aachen University, Aachen, Germany
| | - Inna Pertsovskaya
- Institut d' Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | - Vicky Pliaka
- School of Mechanical Engineering, National Technical University of Athens, Zografou, Greece
| | | | - Gemma Vila
- Institut d' Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | - Pernilla Stridh
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Janina R Behrens
- NeuroCure Clinical Research Center and Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Friedemann Paul
- NeuroCure Clinical Research Center and Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Leonidas G Alexopoulos
- School of Mechanical Engineering, National Technical University of Athens, Zografou, Greece. .,ProtATonce Ltd., Athens, Greece.
| | - Pablo Villoslada
- Institut d' Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain.
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK. .,Joint Research Center for Computational Biomedicine (JRC-COMBINE), Faculty of Medicine, RWTH-Aachen University, Aachen, Germany. .,Institute for Computational Biomedicine, Heidelberg University Hospital and Faculty of Medicine, Heidelberg University, Bioquant, Heidelberg, Germany.
| |
Collapse
|
19
|
B Cells in Neuroinflammation: New Perspectives and Mechanistic Insights. Cells 2021; 10:cells10071605. [PMID: 34206848 PMCID: PMC8305155 DOI: 10.3390/cells10071605] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, the role of B cells in neurological disorders has substantially expanded our perspectives on mechanisms of neuroinflammation. The success of B cell-depleting therapies in patients with CNS diseases such as neuromyelitis optica and multiple sclerosis has highlighted the importance of neuroimmune crosstalk in inflammatory processes. While B cells are essential for the adaptive immune system and antibody production, they are also major contributors of pro- and anti-inflammatory cytokine responses in a number of inflammatory diseases. B cells can contribute to neurological diseases through peripheral immune mechanisms, including production of cytokines and antibodies, or through CNS mechanisms following compartmentalization. Emerging evidence suggests that aberrant pro- or anti-inflammatory B cell populations contribute to neurological processes, including glial activation, which has been implicated in the pathogenesis of several neurodegenerative diseases. In this review, we summarize recent findings on B cell involvement in neuroinflammatory diseases and discuss evidence to support pathogenic immunomodulatory functions of B cells in neurological disorders, highlighting the importance of B cell-directed therapies.
Collapse
|
20
|
Caliskan M, Brown CD, Maranville JC. A catalog of GWAS fine-mapping efforts in autoimmune disease. Am J Hum Genet 2021; 108:549-563. [PMID: 33798443 PMCID: PMC8059376 DOI: 10.1016/j.ajhg.2021.03.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Genome-wide association studies (GWASs) have enabled unbiased identification of genetic loci contributing to common complex diseases. Because GWAS loci often harbor many variants and genes, it remains a major challenge to move from GWASs' statistical associations to the identification of causal variants and genes that underlie these association signals. Researchers have applied many statistical and functional fine-mapping strategies to prioritize genetic variants and genes as potential candidates. There is no gold standard in fine-mapping approaches, but consistent results across different approaches can improve confidence in the fine-mapping findings. Here, we combined text mining with a systematic review and formed a catalog of 85 studies with evidence of fine mapping for at least one autoimmune GWAS locus. Across all fine-mapping studies, we compiled 230 GWAS loci with allelic heterogeneity estimates and predictions of causal variants and trait-relevant genes. These 230 loci included 455 combinations of locus-by-disease association signals with 15 autoimmune diseases. Using these estimates, we assessed the probability of mediating disease risk associations across genes in GWAS loci and identified robust signals of causal disease biology. We predict that this comprehensive catalog of GWAS fine-mapping efforts in autoimmune disease will greatly help distill the plethora of information in the field and inform therapeutic strategies.
Collapse
Affiliation(s)
- Minal Caliskan
- Department of Informatics and Predictive Sciences, Bristol Myers Squibb, Princeton, NJ 08540, USA.
| | - Christopher D Brown
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph C Maranville
- Department of Informatics and Predictive Sciences, Bristol Myers Squibb, Princeton, NJ 08540, USA
| |
Collapse
|
21
|
Cytokines and Transgenic Matrix in Autoimmune Diseases: Similarities and Differences. Biomedicines 2020; 8:biomedicines8120559. [PMID: 33271810 PMCID: PMC7761121 DOI: 10.3390/biomedicines8120559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases are increasingly recognized as disease entities in which dysregulated cytokines contribute to tissue-specific inflammation. In organ-specific and multiorgan autoimmune diseases, the cytokine profiles show some similarities. Despite these similarities, the cytokines have different roles in the pathogenesis of different diseases. Altered levels or action of cytokines can result from changes in cell signaling. This article describes alterations in the JAK-STAT, TGF-β and NF-κB signaling pathways, which are involved in the pathogenesis of multiple sclerosis and systemic lupus erythematosus. There is a special focus on T cells in preclinical models and in patients afflicted with these chronic inflammatory diseases.
Collapse
|
22
|
Akbarian F, Tabatabaiefar MA, Shaygannejad V, Shahpouri MM, Badihian N, Sajjadi R, Dabiri A, Jalilian N, Noori-Daloii MR. Upregulation of MTOR, RPS6KB1, and EIF4EBP1 in the whole blood samples of Iranian patients with multiple sclerosis compared to healthy controls. Metab Brain Dis 2020; 35:1309-1316. [PMID: 32809098 DOI: 10.1007/s11011-020-00590-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 06/23/2020] [Indexed: 12/22/2022]
Abstract
Various genetic and epigenetic mechanisms have been suggested to play roles as the underlying pathophysiology of Multiple Sclerosis (MS). Changes in different parts of the mTOR signaling pathway are among the potential suggested mechanisms based on the specific roles of this pathway in CNS. MTOR, RPS6KB1, and EIFEBP1 genes are among important genes in the mTOR pathway, responsible for the proper function of acting proteins in this signaling pathway. This study aimed to investigate the relative expression levels of these genes in the blood samples of relapsing-remitting MS (RRMS) patients compared to healthy controls. In this case-control study blood samples were collected from 30 newly diagnosed RRMS patients and 30 age and sex-matched healthy controls. mRNA level of MTOR, RPS6KB1, and EIFEBP1 genes were assessed using Real-Time PCR. The expression of MTOR, RPS6KB1, and EIF4EBP1 genes was up regulated in MS patients compared to healthy controls (p < 0.001 for all mentioned genes). Considering gender differences, expression of the mentioned genes was increased among female patients (all P < 0.001). However, no statistically significant changes were observed among male patients. Based on the receiver operating characteristic, MTOR gene had the highest diagnostic value followed by EIF4EBP1 and RPS6KB1 genes in differentiating RRMS patients from controls. In conclusion, we found the simultaneous upregulation of MTOR, RPS6KB1, and EIF4EBP1 genes among RRMS patients. MTOR showed to have the highest diagnostic value compared to other 2 genes in differentiating RRMS patients. Further studies evaluating the importance of these findings from pharmacological and prognostic perspectives are necessary.
Collapse
Affiliation(s)
- Fahimeh Akbarian
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Poursina St., Tehran, 14155-6447, Iran
| | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahid Shaygannejad
- Isfahan Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Negin Badihian
- Isfahan Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roshanak Sajjadi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arezou Dabiri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nazanin Jalilian
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Noori-Daloii
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Poursina St., Tehran, 14155-6447, Iran.
| |
Collapse
|
23
|
The contribution of thymic tolerance to central nervous system autoimmunity. Semin Immunopathol 2020; 43:135-157. [PMID: 33108502 PMCID: PMC7925481 DOI: 10.1007/s00281-020-00822-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022]
Abstract
Autoimmune diseases of the central nervous system (CNS) are associated with high levels of morbidity and economic cost. Research efforts have previously focused on the contribution of the peripheral adaptive and innate immune systems to CNS autoimmunity. However, a failure of thymic negative selection is a necessary step in CNS-reactive T cells escaping into the periphery. Even with defective thymic or peripheral tolerance, the development of CNS inflammation is rare. The reasons underlying this are currently poorly understood. In this review, we examine evidence implicating thymic selection in the pathogenesis of CNS autoimmunity. Animal models suggest that thymic negative selection is an important factor in determining susceptibility to and severity of CNS inflammation. There are indirect clinical data that suggest thymic function is also important in human CNS autoimmune diseases. Specifically, the association between thymoma and paraneoplastic encephalitis and changes in T cell receptor excision circles in multiple sclerosis implicate thymic tolerance in these diseases. We identify potential associations between CNS autoimmunity susceptibility factors and thymic tolerance. The therapeutic manipulation of thymopoiesis has the potential to open up new treatment modalities, but a better understanding of thymic tolerance in CNS autoimmunity is required before this can be realised.
Collapse
|
24
|
N'diaye M, Brauner S, Flytzani S, Kular L, Warnecke A, Adzemovic MZ, Piket E, Min JH, Edwards W, Mela F, Choi HY, Magg V, James T, Linden M, Reichardt HM, Daws MR, van Horssen J, Kockum I, Harris RA, Olsson T, Guerreiro-Cacais AO, Jagodic M. C-type lectin receptors Mcl and Mincle control development of multiple sclerosis-like neuroinflammation. J Clin Invest 2020; 130:838-852. [PMID: 31725411 PMCID: PMC6994148 DOI: 10.1172/jci125857] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Pattern recognition receptors (PRRs) are crucial for responses to infections and tissue damage; however, their role in autoimmunity is less clear. Herein we demonstrate that 2 C-type lectin receptors (CLRs) Mcl and Mincle play an important role in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Congenic rats expressing lower levels of Mcl and Mincle on myeloid cells exhibited a drastic reduction in EAE incidence. In vivo silencing of Mcl and Mincle or blockade of their endogenous ligand SAP130 revealed that these receptors’ expression in the central nervous system is crucial for T cell recruitment and reactivation into a pathogenic Th17/GM-CSF phenotype. Consistent with this, we uncovered MCL- and MINCLE-expressing cells in brain lesions of MS patients and we further found an upregulation of the MCL/MINCLE signaling pathway and an increased response following MCL/MINCLE stimulation in peripheral blood mononuclear cells from MS patients. Together, these data support a role for CLRs in autoimmunity and implicate the MCL/MINCLE pathway as a potential therapeutic target in MS.
Collapse
Affiliation(s)
- Marie N'diaye
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Susanna Brauner
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sevasti Flytzani
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Warnecke
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Milena Z Adzemovic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eliane Piket
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jin-Hong Min
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Will Edwards
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Filia Mela
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hoi Ying Choi
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vera Magg
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tojo James
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magdalena Linden
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Holger M Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Andre O Guerreiro-Cacais
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
A genetic variant of IL17A gene promoter is associated with reduced risk of multiple sclerosis disease in the Iranian population. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Abstract
B cells serve as a key weapon against infectious diseases. They also contribute to multiple autoimmune diseases, including multiple sclerosis (MS) where depletion of B cells is a highly effective therapy. We describe a comprehensive profile of central nervous system (CNS)-specific transcriptional B cell phenotypes in MS at single-cell resolution with paired immune repertoires. We reveal a polyclonal immunoglobulin M (IgM) and IgG1 cerebrospinal fluid B cell expansion polarized toward an inflammatory, memory and plasmablast/plasma cell phenotype, with differential up-regulation of specific proinflammatory pathways. We did not find evidence that CNS B cells harbor a neurotropic virus. These data support the targeting of activated resident B cells in the CNS as a potentially effective strategy for control of treatment-resistant chronic disease. Central nervous system B cells have several potential roles in multiple sclerosis (MS): secretors of proinflammatory cytokines and chemokines, presenters of autoantigens to T cells, producers of pathogenic antibodies, and reservoirs for viruses that trigger demyelination. To interrogate these roles, single-cell RNA sequencing (scRNA-Seq) was performed on paired cerebrospinal fluid (CSF) and blood from subjects with relapsing-remitting MS (RRMS; n = 12), other neurologic diseases (ONDs; n = 1), and healthy controls (HCs; n = 3). Single-cell immunoglobulin sequencing (scIg-Seq) was performed on a subset of these subjects and additional RRMS (n = 4), clinically isolated syndrome (n = 2), and OND (n = 2) subjects. Further, paired CSF and blood B cell subsets (RRMS; n = 7) were isolated using fluorescence activated cell sorting for bulk RNA sequencing (RNA-Seq). Independent analyses across technologies demonstrated that nuclear factor kappa B (NF-κB) and cholesterol biosynthesis pathways were activated, and specific cytokine and chemokine receptors were up-regulated in CSF memory B cells. Further, SMAD/TGF-β1 signaling was down-regulated in CSF plasmablasts/plasma cells. Clonally expanded, somatically hypermutated IgM+ and IgG1+ CSF B cells were associated with inflammation, blood–brain barrier breakdown, and intrathecal Ig synthesis. While we identified memory B cells and plasmablast/plasma cells with highly similar Ig heavy-chain sequences across MS subjects, similarities were also identified with ONDs and HCs. No viral transcripts, including from Epstein–Barr virus, were detected. Our findings support the hypothesis that in MS, CSF B cells are driven to an inflammatory and clonally expanded memory and plasmablast/plasma cell phenotype.
Collapse
|
27
|
Mortales CL, Lee SU, Manousadjian A, Hayama KL, Demetriou M. N-Glycan Branching Decouples B Cell Innate and Adaptive Immunity to Control Inflammatory Demyelination. iScience 2020; 23:101380. [PMID: 32745987 PMCID: PMC7398982 DOI: 10.1016/j.isci.2020.101380] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/10/2020] [Accepted: 07/14/2020] [Indexed: 11/21/2022] Open
Abstract
B cell depletion potently reduces episodes of inflammatory demyelination in multiple sclerosis (MS), predominantly through loss of innate rather than adaptive immunity. However, molecular mechanisms controlling innate versus adaptive B cell function are poorly understood. N-glycan branching, via interactions with galectins, controls endocytosis and signaling of cell surface receptors to control cell function. Here we report that N-glycan branching in B cells dose dependently reduces pro-inflammatory innate responses by titrating decreases in Toll-like receptor-4 (TLR4) and TLR2 surface expression via endocytosis. In contrast, a minimal level of N-glycan branching maximizes surface retention of the B cell receptor (BCR) and the CD19 co-receptor to promote adaptive immunity. Branched N-glycans inhibit antigen presentation by B cells to reduce T helper cell-17 (TH17)/TH1 differentiation and inflammatory demyelination in mice. Thus, N-glycan branching negatively regulates B cell innate function while promoting/maintaining adaptive immunity via BCR, providing an attractive therapeutic target for MS.
Collapse
Affiliation(s)
- Christie-Lynn Mortales
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92617, USA
| | - Sung-Uk Lee
- Department of Neurology, University of California, Irvine, CA 92617, USA
| | - Armen Manousadjian
- Department of Neurology, University of California, Irvine, CA 92617, USA
| | - Ken L Hayama
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92617, USA
| | - Michael Demetriou
- Department of Neurology, University of California, Irvine, CA 92617, USA; Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92617, USA.
| |
Collapse
|
28
|
Yan J, McCombe PA, Pender MP, Greer JM. Reduced IκB-α Protein Levels in Peripheral Blood Cells of Patients with Multiple Sclerosis-A Possible Cause of Constitutive NF-κB Activation. J Clin Med 2020; 9:jcm9082534. [PMID: 32781504 PMCID: PMC7465818 DOI: 10.3390/jcm9082534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 11/16/2022] Open
Abstract
NF-κB signaling pathways are dysregulated in both the central nervous system (CNS) and peripheral blood cells in multiple sclerosis (MS), but the cause of this is unknown. We have recently reported that peripheral blood mononuclear cells (PBMC) of patients with MS have increased constitutive activation and translocation of the transcription factor NF-κB to the nucleus compared to healthy subjects. NF-κB can be activated through either canonical or non-canonical pathways. In the canonical pathway, activation of NF-κB is normally negatively regulated by the inhibitor IκB. We therefore hypothesized that the increased activation of NF-κB could be caused by reduced IκB-α in the cells of patients with MS, possibly due to increased activity of the IκB kinase (IKK) complex, which regulates IκB-α. Alternatively, changes to the activity of key molecules in the non-canonical pathway, such as IKKα, could also lead to increased NF-κB activation. We therefore used Western blotting to detect IκB-α levels and ELISA to investigate NF-κB DNA binding activity and phosphorylation of IKKα and IKKβ in samples from PBMC of MS patients and controls. The level of full-length IκB-α protein in the cytosolic fraction of PBMC of MS patients was significantly reduced compared to healthy subjects, with significantly more evidence of multiple low molecular weight putative degradation products of IκB-α present in MS patients compared to healthy subjects. Conversely, the level of NF-κB DNA binding activity was increased in whole cell lysates from MS patients. Both IKKα and IKKβ showed increased overall activity in MS compared to healthy subjects, although not all of the MS patients showed increased activity compared to the healthy subjects, suggesting that there may be several different mechanisms underlying the constitutive activation of NF-κB in MS. Taken together, these findings suggest that there may be multiple points at which the NF-κB pathway is dysregulated in MS and that decreased levels of the full-length IκB-α protein are a major component in this.
Collapse
Affiliation(s)
- Jun Yan
- UQ Centre for Clinical Research, The University of Queensland Centre for Clinical Research, Brisbane, QLD 4029, Australia; (J.Y.); (P.A.M.)
| | - Pamela A. McCombe
- UQ Centre for Clinical Research, The University of Queensland Centre for Clinical Research, Brisbane, QLD 4029, Australia; (J.Y.); (P.A.M.)
- Wesley Medical Research, The Wesley Hospital, Auchenflower, QLD 4066, Australia
| | - Michael P. Pender
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia;
- Department of Neurology, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia
| | - Judith M. Greer
- UQ Centre for Clinical Research, The University of Queensland Centre for Clinical Research, Brisbane, QLD 4029, Australia; (J.Y.); (P.A.M.)
- Wesley Medical Research, The Wesley Hospital, Auchenflower, QLD 4066, Australia
- Correspondence: ; Tel.: +(61)-07-3346-6018
| |
Collapse
|
29
|
Li T, Li X, Zamani A, Wang W, Lee CN, Li M, Luo G, Eiler E, Sun H, Ghosh S, Jin J, Murali R, Ruan Q, Shi W, Chen YH. c-Rel Is a Myeloid Checkpoint for Cancer Immunotherapy. ACTA ACUST UNITED AC 2020; 1:507-517. [PMID: 33458695 DOI: 10.1038/s43018-020-0061-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Immunotherapy that targets lymphoid cell checkpoints holds great promise for curing cancer. However, a majority of cancer patients do not respond to this form of therapy. In addition to lymphoid cells, myeloid cells play essential roles in controlling immunity to cancer. Whether myeloid checkpoints exist that can be targeted to treat cancer is not well established. Here we show that c-Rel, a member of the nuclear factor (NF)-B family, specified the generation of myeloid-derived suppressor cells (MDSCs) by selectively turning on pro-tumoral genes while switching off anti-tumoral genes through a c-Rel enhanceosome. c-Rel deficiency in myeloid cells markedly inhibited cancer growth in mice, and pharmaceutical inhibition of c-Rel had the same effect. Combination therapy that blocked both c-Rel and the lymphoid checkpoint protein PD1 was more effective in treating cancer than blocking either alone. Thus, c-Rel is a myeloid checkpoint that can be targeted for treating cancer.
Collapse
Affiliation(s)
- Ting Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Contributed equally to this work
| | - Xinyuan Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Contributed equally to this work
| | - Ali Zamani
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chin-Nien Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mingyue Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - George Luo
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily Eiler
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Honghong Sun
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ramachandran Murali
- Department of Biomedical Sciences, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Qingguo Ruan
- Shandong Eye Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Weiyun Shi
- Shandong Eye Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
30
|
van Langelaar J, Rijvers L, Smolders J, van Luijn MM. B and T Cells Driving Multiple Sclerosis: Identity, Mechanisms and Potential Triggers. Front Immunol 2020; 11:760. [PMID: 32457742 PMCID: PMC7225320 DOI: 10.3389/fimmu.2020.00760] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 04/03/2020] [Indexed: 12/25/2022] Open
Abstract
Historically, multiple sclerosis (MS) has been viewed as being primarily driven by T cells. However, the effective use of anti-CD20 treatment now also reveals an important role for B cells in MS patients. The results from this treatment put forward T-cell activation rather than antibody production by B cells as a driving force behind MS. The main question of how their interaction provokes both B and T cells to infiltrate the CNS and cause local pathology remains to be answered. In this review, we highlight key pathogenic events involving B and T cells that most likely contribute to the pathogenesis of MS. These include (1) peripheral escape of B cells from T cell-mediated control, (2) interaction of pathogenic B and T cells in secondary lymph nodes, and (3) reactivation of B and T cells accumulating in the CNS. We will focus on the functional programs of CNS-infiltrating lymphocyte subsets in MS patients and discuss how these are defined by mechanisms such as antigen presentation, co-stimulation and cytokine production in the periphery. Furthermore, the potential impact of genetic variants and viral triggers on candidate subsets will be debated in the context of MS.
Collapse
Affiliation(s)
- Jamie van Langelaar
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Liza Rijvers
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Joost Smolders
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Marvin M. van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
31
|
Fazia T, Nova A, Gentilini D, Beecham A, Piras M, Saddi V, Ticca A, Bitti P, McCauley JL, Berzuini C, Bernardinelli L. Investigating the Causal Effect of Brain Expression of CCL2, NFKB1, MAPK14, TNFRSF1A, CXCL10 Genes on Multiple Sclerosis: A Two-Sample Mendelian Randomization Approach. Front Bioeng Biotechnol 2020; 8:397. [PMID: 32432099 PMCID: PMC7216783 DOI: 10.3389/fbioe.2020.00397] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/08/2020] [Indexed: 12/17/2022] Open
Abstract
Multiple Sclerosis (MS) exhibits considerable heterogeneity in phenotypic expression, course, prognosis and response to therapy. This suggests this disease involves multiple, as yet poorly understood, causal mechanisms. In this work we assessed the possible causal link between gene expression level of five selected genes related to the pro-inflammatory NF-κB signaling pathway (i.e., CCL2, NFKB1, MAPK14, TNFRSF1A, CXCL10) in ten different brain tissues (i.e., cerebellum, frontal cortex, hippocampus, medulla, occipital cortex, putamen, substantia nigra, thalamus, temporal cortex and intralobular white matter) and MS. We adopted a two-stage Mendelian Randomization (MR) approach for the estimation of the causal effects of interest, based on summary-level data from 20 multiplex Sardinian families and data provided by the United Kingdom Brain Expression Consortium (UKBEC). Through Radial-MR and Cochrane's Q statistics we identified and removed genetic variants which are most likely to be invalid instruments. To estimate the total causal effect, univariable MR was carried out separately for each gene and brain region. We used Inverse-Variance Weighted estimator (IVW) as main analysis and MR-Egger Regression estimator (MR-ER) and Weighted Median Estimator (WME) as sensitivity analysis. As these genes belong to the same pathway and thus they can be closely related, we also estimated their direct causal effects by applying IVW and MR-ER within a multivariable MR (MVMR) approach using set of genetic instruments specific and common (composite) to each multiple exposures represented by the expression of the candidate genes. Univariate MR analysis showed a significant positive total causal effect for CCL2 and NFKB1 respectively in medulla and cerebellum. MVMR showed a direct positive causal effect for NFKB1 and TNFRSF1A, and a direct negative causal effect for CCL2 in cerebellum; while in medulla we observed a direct positive causal effect for CCL2. Since in general we observed a different magnitude for the gene specific causal effect we hypothesize that in cerebellum and medulla the effect of each gene expression is direct but also mediated by the others. These results confirm the importance of the involvement of NF-κB signaling pathway in brain tissue for the development of the disease and improve our understanding in the pathogenesis of MS.
Collapse
Affiliation(s)
- Teresa Fazia
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Andrea Nova
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Davide Gentilini
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, Milan, Italy
- Molecular Biology Laboratory, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Ashley Beecham
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL, United States
| | - Marialuisa Piras
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Valeria Saddi
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Anna Ticca
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Pierpaolo Bitti
- Centro di Tipizzazione Tissutale, S.I.T., Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Jacob L. McCauley
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL, United States
| | - Carlo Berzuini
- Centre for Biostatistics, University of Manchester, Manchester, United Kingdom
| | - Luisa Bernardinelli
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
32
|
Sun H, Lin M, Zamani A, Goldsmith JR, Boggs AE, Li M, Lee CN, Chen X, Li X, Li T, Dorrity BL, Li N, Lou Y, Shi S, Wang W, Chen YH. The TIPE Molecular Pilot That Directs Lymphocyte Migration in Health and Inflammation. Sci Rep 2020; 10:6617. [PMID: 32313148 PMCID: PMC7170861 DOI: 10.1038/s41598-020-63629-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Lymphocytes are some of the most motile cells of vertebrates, constantly navigating through various organ systems. Their specific positioning in the body is delicately controlled by site-specific directional cues such as chemokines. While it has long been suspected that an intrinsic molecular pilot, akin to a ship's pilot, guides lymphocyte navigation, the nature of this pilot is unknown. Here we show that the TIPE (TNF-α-induced protein 8-like) family of proteins pilot lymphocytes by steering them toward chemokines. TIPE proteins are carriers of lipid second messengers. They mediate chemokine-induced local generation of phosphoinositide second messengers, but inhibit global activation of the small GTPase Rac. TIPE-deficient T lymphocytes are completely pilot-less: they are unable to migrate toward chemokines despite their normal ability to move randomly. As a consequence, TIPE-deficient mice have a marked defect in positioning their T lymphocytes to various tissues, both at the steady-state and during inflammation. Thus, TIPE proteins pilot lymphocytes during migration and may be targeted for the treatment of lymphocyte-related disorders.
Collapse
Affiliation(s)
- Honghong Sun
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mei Lin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Zamani
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason R Goldsmith
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda E Boggs
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mingyue Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chin-Nien Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xu Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xinyuan Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ting Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brigid L Dorrity
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ning Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yunwei Lou
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Songlin Shi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Zhou Y, Cui C, Ma X, Luo W, Zheng SG, Qiu W. Nuclear Factor κB (NF-κB)-Mediated Inflammation in Multiple Sclerosis. Front Immunol 2020; 11:391. [PMID: 32265906 PMCID: PMC7105607 DOI: 10.3389/fimmu.2020.00391] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/19/2020] [Indexed: 12/18/2022] Open
Abstract
The nuclear factor κB (NF-κB) signaling cascade has been implicating in a broad range of biological processes, including inflammation, cell proliferation, differentiation, and apoptosis. The past three decades have witnessed a great progress in understanding the impact of aberrant NF-κB regulation on human autoimmune and inflammatory disorders. In this review, we discuss how aberrant NF-κB activation contributes to multiple sclerosis, a typical inflammatory demyelinating disease of the central nervous system, and its involvement in developing potential therapeutic targets.
Collapse
Affiliation(s)
- Yifan Zhou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunping Cui
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyu Ma
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenjing Luo
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Song Guo Zheng
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
34
|
Milo R. Ofatumumab – A Potential Subcutaneous B-cell Therapy for Relapsing Multiple Sclerosis. ACTA ACUST UNITED AC 2020. [DOI: 10.17925/enr.2020.15.1.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
35
|
Ciuffoli V, Lena AM, Gambacurta A, Melino G, Candi E. Myoblasts rely on TAp63 to control basal mitochondria respiration. Aging (Albany NY) 2019; 10:3558-3573. [PMID: 30487319 PMCID: PMC6286837 DOI: 10.18632/aging.101668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022]
Abstract
p53, with its family members p63 and p73, have been shown to promote myoblast differentiation by regulation of the function of the retinoblastoma protein and by direct activation of p21Cip/Waf1 and p57Kip2, promoting cell cycle exit. In previous studies, we have demonstrated that the TAp63γ isoform is the only member of the p53 family that accumulates during in vitro myoblasts differentiation, and that its silencing led to delay in myotube fusion. To better dissect the role of TAp63γ in myoblast physiology, we have generated both sh-p63 and Tet-On inducible TAp63γ clones. Gene array analysis of sh-p63 C2C7 clones showed a significant modulation of genes involved in proliferation and cellular metabolism. Indeed, we found that sh-p63 C2C7 myoblasts present a higher proliferation rate and that, conversely, TAp63γ ectopic expression decreases myoblasts proliferation, indicating that TAp63γ specifically contributes to myoblasts proliferation, independently of p53 and p73. In addition, sh-p63 cells have a defect in mitochondria respiration highlighted by a reduction in spare respiratory capacity and a decrease in complex I, IV protein levels. These results demonstrated that, beside contributing to cell cycle exit, TAp63γ participates to myoblasts metabolism control.
Collapse
Affiliation(s)
- Veronica Ciuffoli
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Alessandra Gambacurta
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy.,MRC-Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Eleonora Candi
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy.,IDI-IRCCS, Biochemistry laboratory, Rome, Italy
| |
Collapse
|
36
|
Frezza V, Fierro C, Gatti E, Peschiaroli A, Lena AM, Petruzzelli MA, Candi E, Anemona L, Mauriello A, Pelicci PG, Melino G, Bernassola F. ΔNp63 promotes IGF1 signalling through IRS1 in squamous cell carcinoma. Aging (Albany NY) 2019; 10:4224-4240. [PMID: 30594912 PMCID: PMC6326668 DOI: 10.18632/aging.101725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
Accumulating evidence has proved that deregulation of ΔNp63 expression plays an oncogenic role in head and neck squamous cell carcinomas (HNSCCs). Besides p63, the type 1-insulin-like growth factor (IGF) signalling pathway has been implicated in HNSCC development and progression. Most insulin/IGF1 signalling converges intracellularly onto the protein adaptor insulin receptor substrate-1 (IRS-1) that transmits signals from the receptor to downstream effectors, including the PI3K/AKT and the MAPK kinase pathways, which, ultimately, promote proliferation, invasion, and cell survival. Here we report that p63 directly controls IRS1 transcription and cellular abundance and fosters the PI3K/AKT and MAPK downstream signalling pathways. Inactivation of ΔNp63 expression indeed reduces tumour cell responsiveness to IGF1 stimulation, and inhibits the growth potential of HNSCC cells. In addition, a positive correlation was observed between p63 and IRS1 expression in human HNSCC tissue arrays and in publicly available gene expression data. Our findings indicate that aberrant expression of ΔNp63 in HNSSC may act as an oncogenic stimulus by altering the IGF signalling pathway.
Collapse
Affiliation(s)
- Valentina Frezza
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Claudia Fierro
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Elena Gatti
- Department of Experimental Oncology European Institute of Oncology, Milan 20139, Italy
| | - Angelo Peschiaroli
- National Research Council of Italy Institute of Translational Pharmacology (IFT-CNR), Rome 00133, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | | | - Eleonora Candi
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy.,Istituto Dermopatico dell'Immacolata, IRCCS,, Rome 00163, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology European Institute of Oncology, Milan 20139, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy.,Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, UK
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| |
Collapse
|
37
|
Abstract
Emerging data point to important contributions of both autoimmune inflammation and progressive degeneration in the pathophysiology of multiple sclerosis (MS). Unfortunately, after decades of intensive investigation, the fundamental cause remains unknown. A large body of research on the immunobiology of MS has resulted in a variety of anti-inflammatory therapies that are highly effective at reducing brain inflammation and clinical/radiological relapses. However, despite potent suppression of inflammation, benefit in the more important and disabling progressive phase is extremely limited; thus, progressive MS has emerged as the greatest challenge for the MS research and clinical communities. Data obtained over the years point to a complex interplay between environment (e.g., the near-absolute requirement of Epstein-Barr virus exposure), immunogenetics (strong associations with a large number of immune genes), and an ever more convincing role of an underlying degenerative process resulting in demyelination (in both white and grey matter regions), axonal and neuro-synaptic injury, and a persistent innate inflammatory response with a seemingly diminishing role of T cell-mediated autoimmunity as the disease progresses. Together, these observations point toward a primary degenerative process, one whose cause remains unknown but one that entrains a nearly ubiquitous secondary autoimmune response, as a likely sequence of events underpinning this disease. Here, we briefly review what is known about the potential pathophysiological mechanisms, focus on progressive MS, and discuss the two main hypotheses of MS pathogenesis that are the topic of vigorous debate in the field: whether primary autoimmunity or degeneration lies at the foundation. Unravelling this controversy will be critically important for developing effective new therapies for the most disabling later phases of this disease.
Collapse
Affiliation(s)
- Peter K. Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Medicine University of Calgary, Calgary, Alberta, Canada
| | - Shigeki Tsutsui
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Medicine University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
38
|
Abstract
Increasing evidence suggests that B cells contribute both to the regulation of normal autoimmunity and to the pathogenesis of immune mediated diseases, including multiple sclerosis (MS). B cells in MS are skewed toward a pro-inflammatory profile, and contribute to MS pathogenesis by antibody production, antigen presentation, T cells stimulation and activation, driving autoproliferation of brain-homing autoreactive CD4+ T cells, production of pro-inflammatory cytokines, and formation of ectopic meningeal germinal centers that drive cortical pathology and contribute to neurological disability. The recent interest in the key role of B cells in MS has been evoked by the profound anti-inflammatory effects of rituximab, a chimeric monoclonal antibody (mAb) targeting the B cell surface marker CD20, observed in relapsing-remitting MS. This has been reaffirmed by clinical trials with less immunogenic and more potent B cell-depleting mAbs targeting CD20 – ocrelizumab, ofatumumab and ublituximab. Ocrelizumab is also the first disease-modifying drug that has shown efficacy in primary-progressive MS, and is currently approved for both indications. Another promising approach is the inhibition of Bruton's tyrosine kinase, a key enzyme that mediates B cell activation and survival, by agents such as evobrutinib. On the other hand, targeting B cell cytokines with the fusion protein atacicept increased MS activity, highlighting the complex and not fully understood role of B cells and humoral immunity in MS. Finally, all other approved therapies for MS, some of which have been designed to target T cells, have some effects on the frequency, phenotype, or homing of B cells, which may contribute to their therapeutic activity.
Collapse
Affiliation(s)
- Ron Milo
- Ron Milo, Department of Neurology, Barzilai Medical Center, Ha-Histadrut St 2, Ashkelon 7308604, Israel,
| |
Collapse
|
39
|
Milo R. Therapies for multiple sclerosis targeting B cells. Croat Med J 2019; 60:87-98. [PMID: 31044580 PMCID: PMC6509632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 04/14/2019] [Indexed: 10/12/2023] Open
Abstract
Increasing evidence suggests that B cells contribute both to the regulation of normal autoimmunity and to the pathogenesis of immune mediated diseases, including multiple sclerosis (MS). B cells in MS are skewed toward a pro-inflammatory profile, and contribute to MS pathogenesis by antibody production, antigen presentation, T cells stimulation and activation, driving autoproliferation of brain-homing autoreactive CD4+ T cells, production of pro-inflammatory cytokines, and formation of ectopic meningeal germinal centers that drive cortical pathology and contribute to neurological disability. The recent interest in the key role of B cells in MS has been evoked by the profound anti-inflammatory effects of rituximab, a chimeric monoclonal antibody (mAb) targeting the B cell surface marker CD20, observed in relapsing-remitting MS. This has been reaffirmed by clinical trials with less immunogenic and more potent B cell-depleting mAbs targeting CD20 - ocrelizumab, ofatumumab and ublituximab. Ocrelizumab is also the first disease-modifying drug that has shown efficacy in primary-progressive MS, and is currently approved for both indications. Another promising approach is the inhibition of Bruton's tyrosine kinase, a key enzyme that mediates B cell activation and survival, by agents such as evobrutinib. On the other hand, targeting B cell cytokines with the fusion protein atacicept increased MS activity, highlighting the complex and not fully understood role of B cells and humoral immunity in MS. Finally, all other approved therapies for MS, some of which have been designed to target T cells, have some effects on the frequency, phenotype, or homing of B cells, which may contribute to their therapeutic activity.
Collapse
Affiliation(s)
- Ron Milo
- Ron Milo, Department of Neurology, Barzilai Medical Center, Ha-Histadrut St 2, Ashkelon 7308604, Israel,
| |
Collapse
|
40
|
Webb LM, Narvaez Miranda J, Amici SA, Sengupta S, Nagy G, Guerau-de-Arellano M. NF-κB/mTOR/MYC Axis Drives PRMT5 Protein Induction After T Cell Activation via Transcriptional and Non-transcriptional Mechanisms. Front Immunol 2019; 10:524. [PMID: 30941147 PMCID: PMC6433977 DOI: 10.3389/fimmu.2019.00524] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/26/2019] [Indexed: 12/17/2022] Open
Abstract
Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) mediated by CD4+ T cells and modeled via experimental autoimmune encephalomyelitis (EAE). Inhibition of PRMT5, the major Type II arginine methyltransferase, suppresses pathogenic T cell responses and EAE. PRMT5 is transiently induced in proliferating memory inflammatory Th1 cells and during EAE. However, the mechanisms driving PRMT5 protein induction and repression as T cells expand and return to resting is currently unknown. Here, we used naive mouse and memory mouse and human Th1/Th2 cells as models to identify mechanisms controlling PRMT5 protein expression in initial and recall T cell activation. Initial activation of naive mouse T cells resulted in NF-κB-dependent transient Prmt5 transcription and NF-κB, mTOR and MYC-dependent PRMT5 protein induction. In murine memory Th cells, transcription and miRNA loss supported PRMT5 induction to a lesser extent than in naive T cells. In contrast, NF-κB/MYC/mTOR-dependent non-transcriptional PRMT5 induction played a major role. These results highlight the importance of the NF-κB/mTOR/MYC axis in PRMT5-driven pathogenic T cell expansion and may guide targeted therapeutic strategies for MS.
Collapse
Affiliation(s)
- Lindsay M Webb
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Janiret Narvaez Miranda
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Stephanie A Amici
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Shouvonik Sengupta
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Gregory Nagy
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Mireia Guerau-de-Arellano
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
41
|
Delgado ME, Brunner T. The many faces of tumor necrosis factor signaling in the intestinal epithelium. Genes Immun 2019; 20:609-626. [DOI: 10.1038/s41435-019-0057-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/26/2018] [Indexed: 01/15/2023]
|
42
|
Rojas M, Restrepo-Jiménez P, Monsalve DM, Pacheco Y, Acosta-Ampudia Y, Ramírez-Santana C, Leung PS, Ansari AA, Gershwin ME, Anaya JM. Molecular mimicry and autoimmunity. J Autoimmun 2018; 95:100-123. [DOI: 10.1016/j.jaut.2018.10.012] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/12/2018] [Accepted: 10/16/2018] [Indexed: 12/15/2022]
|
43
|
Lacher SM, Thurm C, Distler U, Mohebiany AN, Israel N, Kitic M, Ebering A, Tang Y, Klein M, Wabnitz GH, Wanke F, Samstag Y, Bopp T, Kurschus FC, Simeoni L, Tenzer S, Waisman A. NF-κB inducing kinase (NIK) is an essential post-transcriptional regulator of T-cell activation affecting F-actin dynamics and TCR signaling. J Autoimmun 2018; 94:110-121. [PMID: 30061013 DOI: 10.1016/j.jaut.2018.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 11/16/2022]
Abstract
NF-κB inducing kinase (NIK) is the key protein of the non-canonical NF-κB pathway and is important for the development of lymph nodes and other secondary immune organs. We elucidated the specific role of NIK in T cells using T-cell specific NIK-deficient (NIKΔT) mice. Despite showing normal development of lymphoid organs, NIKΔT mice were resistant to induction of CNS autoimmunity. T cells from NIKΔT mice were deficient in late priming, failed to up-regulate T-bet and to transmigrate into the CNS. Proteomic analysis of activated NIK-/- T cells showed de-regulated expression of proteins involved in the formation of the immunological synapse: in particular, proteins involved in cytoskeleton dynamics. In line with this we found that NIK-deficient T cells were hampered in phosphorylation of Zap70, LAT, AKT, ERK1/2 and PLCγ upon TCR engagement. Hence, our data disclose a hitherto unknown function of NIK in T-cell priming and differentiation.
Collapse
MESH Headings
- Actins/genetics
- Actins/immunology
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/immunology
- Animals
- Central Nervous System/immunology
- Central Nervous System/pathology
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Expression Profiling
- Gene Expression Regulation
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphocyte Activation
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/immunology
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/immunology
- Myelin-Oligodendrocyte Glycoprotein/administration & dosage
- Peptide Fragments/administration & dosage
- Phospholipase C gamma/genetics
- Phospholipase C gamma/immunology
- Phosphoproteins/genetics
- Phosphoproteins/immunology
- Primary Cell Culture
- Protein Serine-Threonine Kinases/deficiency
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/immunology
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction
- Spleen/immunology
- Spleen/pathology
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- ZAP-70 Protein-Tyrosine Kinase/genetics
- ZAP-70 Protein-Tyrosine Kinase/immunology
- NF-kappaB-Inducing Kinase
Collapse
Affiliation(s)
- Sonja M Lacher
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christoph Thurm
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation, Otto von Guericke University, Magdeburg, Germany
| | - Ute Distler
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Alma N Mohebiany
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nicole Israel
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Maja Kitic
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anna Ebering
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Yilang Tang
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Guido H Wabnitz
- Institute of Immunology, Section Molecular Immunology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Florian Wanke
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Florian C Kurschus
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luca Simeoni
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation, Otto von Guericke University, Magdeburg, Germany
| | - Stefan Tenzer
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| |
Collapse
|
44
|
Raschellà G, Melino G, Gambacurta A. Cell death in cancer in the era of precision medicine. Genes Immun 2018; 20:529-538. [PMID: 30341419 DOI: 10.1038/s41435-018-0048-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 09/26/2018] [Accepted: 10/01/2018] [Indexed: 12/11/2022]
Abstract
Tumors constitute a large class of diseases that affect different organs and cell lineages. The molecular characterization of cancers of a given type has revealed an extraordinary heterogeneity in terms of genetic alterations and DNA mutations; heterogeneity that is further highlighted by single-cell DNA sequencing of individual patients. To address these issues, drugs that specifically target genes or altered pathways in cancer cells are continuously developed. Indeed, the genetic fingerprint of individual tumors can direct the modern therapeutic approaches to selectively hit the tumor cells while sparing the healthy ones. In this context, the concept of precision medicine finds a vast field of application. In this review, we will briefly list some classes of target drugs (Bcl-2 family modulators, Tyrosine Kinase modulators, PARP inhibitors, and growth factors inhibitors) and discuss the application of immunotherapy in tumors (T cell-mediated immunotherapy and CAR-T cells) that in recent years has drastically changed the prognostic outlook of aggressive cancers. We will also consider how apoptosis could represent a primary end point in modern cancer therapy and how "classic" chemotherapeutic drugs that induce apoptosis are still utilized in therapeutic schedules that involve the use of target drugs or immunotherapy to optimize the antitumor response.
Collapse
Affiliation(s)
- Giuseppe Raschellà
- ENEA Research Center Casaccia, Laboratory of Biosafety and Risk Assessment, Via Anguillarese, 301, 00123, Rome, Italy.
| | - Gerry Melino
- Department of Experimental Medicine TOR, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.,Medical Research Council, Toxicology Unit, Hodgkin Building, University of Cambridge, Leicester, LE1 9HN, UK
| | - Alessandra Gambacurta
- Department of Experimental Medicine TOR, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| |
Collapse
|
45
|
Yan J, Winterford CM, Catts VS, Pat BK, Pender MP, McCombe PA, Greer JM. Increased constitutive activation of NF-κB p65 (RelA) in peripheral blood cells of patients with progressive multiple sclerosis. J Neuroimmunol 2018; 320:111-116. [DOI: 10.1016/j.jneuroim.2018.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/06/2018] [Accepted: 04/02/2018] [Indexed: 12/17/2022]
|
46
|
Mills EA, Mao-Draayer Y. Understanding Progressive Multifocal Leukoencephalopathy Risk in Multiple Sclerosis Patients Treated with Immunomodulatory Therapies: A Bird's Eye View. Front Immunol 2018; 9:138. [PMID: 29456537 PMCID: PMC5801425 DOI: 10.3389/fimmu.2018.00138] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/16/2018] [Indexed: 12/14/2022] Open
Abstract
The increased use of newer potent immunomodulatory therapies for multiple sclerosis (MS), including natalizumab, fingolimod, and dimethyl fumarate, has expanded the patient population at risk for developing progressive multifocal leukoencephalopathy (PML). These MS therapies shift the profile of lymphocytes within the central nervous system (CNS) leading to increased anti-inflammatory subsets and decreased immunosurveillance. Similar to MS, PML is a demyelinating disease of the CNS, but it is caused by the JC virus. The manifestation of PML requires the presence of an active, genetically rearranged form of the JC virus within CNS glial cells, coupled with the loss of appropriate JC virus-specific immune responses. The reliability of metrics used to predict risk for PML could be improved if all three components, i.e., viral genetic strain, localization, and host immune function, were taken into account. Advances in our understanding of the critical lymphocyte subpopulation changes induced by these MS therapies and ability to detect viral mutation and reactivation will facilitate efforts to develop these metrics.
Collapse
Affiliation(s)
- Elizabeth A Mills
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States.,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
47
|
Choriodecidual leukocytes display a unique gene expression signature in spontaneous labor at term. Genes Immun 2018; 20:56-68. [PMID: 29362510 PMCID: PMC6358585 DOI: 10.1038/s41435-017-0010-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/01/2017] [Accepted: 11/16/2017] [Indexed: 12/11/2022]
Abstract
Prior to and during the process of human labor, maternal circulating leukocytes infiltrate the maternal-fetal interface (choriodecidua) and become activated resembling choriodecidual leukocytes. Since, there is no evidence comparing maternal circulating and choriodecidual leukocytes, herein, we characterized their transcriptome and explored the biological processes enriched in choriodecidual leukocytes. From women undergoing spontaneous term labor we isolated circulating and choriodecidual leukocytes, performed microarray analysis (n = 5) and qRT-PCR validation (n = 9) and interaction network analysis with up-regulated genes. We found 270 genes up-regulated and only 17 genes down-regulated in choriodecidual leukocytes compared to maternal circulating leukocytes. The most up-regulated genes were CCL18, GPNMB, SEPP1, FN1, RNASE1, SPP1, C1QC, and PLTP. The biological processes enriched in choriodecidual leukocytes were cell migration and regulation of immune response, chemotaxis, and humoral immune responses. Our results show striking differences between the transcriptome of choriodecidual and maternal circulating leukocytes. Choriodecidual leukocytes are enriched in immune mediators implicated in the spontaneous process of labor at term.
Collapse
|
48
|
Constitutive expression of NF-κB inducing kinase in regulatory T cells impairs suppressive function and promotes instability and pro-inflammatory cytokine production. Sci Rep 2017; 7:14779. [PMID: 29116141 PMCID: PMC5677020 DOI: 10.1038/s41598-017-14965-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 10/16/2017] [Indexed: 12/22/2022] Open
Abstract
CD4+Foxp3+ regulatory T cells (Tregs) are indispensable negative regulators of immune responses. To understand Treg biology in health and disease, it is critical to elucidate factors that affect Treg homeostasis and suppressive function. Tregs express several costimulatory TNF receptor family members that activate non-canonical NF-κB via accumulation of NF-κB inducing kinase (NIK). We previously showed that constitutive NIK expression in all T cells causes fatal multi-organ autoimmunity associated with hyperactive conventional T cell responses and poor Treg-mediated suppression. Here, we show that constitutive NIK expression that is restricted to Tregs via a Cre-inducible transgene causes an autoimmune syndrome. We found that constitutive NIK expression decreased expression of numerous Treg signature genes and microRNAs involved in Treg homeostasis and suppressive phenotype. NIK transgenic Tregs competed poorly with WT Tregs in vivo and produced pro-inflammatory cytokines upon stimulation. Lineage tracing experiments revealed accumulation of ex-Foxp3+ T cells in mice expressing NIK constitutively in Tregs, and these former Tregs produced copious IFNγ and IL-2. Our data indicate that under inflammatory conditions in which NIK is activated, Tregs may lose suppressive function and may actively contribute to inflammation.
Collapse
|
49
|
Vavougios GD, Zarogiannis SG, Krogfelt KA, Gourgoulianis K, Mitsikostas DD, Hadjigeorgiou G. Novel candidate genes of the PARK7 interactome as mediators of apoptosis and acetylation in multiple sclerosis: An in silico analysis. Mult Scler Relat Disord 2017; 19:8-14. [PMID: 29100048 DOI: 10.1016/j.msard.2017.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/30/2017] [Accepted: 10/20/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND currently only 4 studies have explored the potential role of PARK7's dysregulation in MS pathophysiology Currently, no study has evaluated the potential role of the PARK7 interactome in MS. OBJECTIVE The aim of our study was to assess the differential expression of PARK7 mRNA in peripheral blood mononuclears (PBMCs) donated from MS versus healthy patients using data mining techniques. METHODS The PARK7 interactome data from the GDS3920 profile were scrutinized for differentially expressed genes (DEGs); Gene Enrichment Analysis (GEA) was used to detect significantly enriched biological functions. RESULTS 27 differentially expressed genes in the MS dataset were detected; 12 of these (NDUFA4, UBA2, TDP2, NPM1, NDUFS3, SUMO1, PIAS2, KIAA0101, RBBP4, NONO, RBBP7 AND HSPA4) are reported for the first time in MS. Stepwise Linear Discriminant Function Analysis constructed a predictive model (Wilk's λ = 0.176, χ2 = 45.204, p = 1.5275e-10) with 2 variables (TIDP2, RBBP4) that achieved 96.6% accuracy when discriminating between patients and controls. Gene Enrichment Analysis revealed that induction and regulation of programmed / intrinsic cell death represented the most salient Gene Ontology annotations. Cross-validation on systemic lupus erythematosus and ischemic stroke datasets revealed that these functions are unique to the MS dataset. CONCLUSIONS Based on our results, novel potential target genes are revealed; these differentially expressed genes regulate epigenetic and apoptotic pathways that may further elucidate underlying mechanisms of autorreactivity in MS.
Collapse
Affiliation(s)
- George D Vavougios
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, BIOPOLIS, Larissa 41110, Greece.
| | - Sotirios G Zarogiannis
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, BIOPOLIS, Larissa 41110, Greece; Department of Physiology, Faculty of Medicine, University of Thessaly, BIOPOLIS, Larissa 41110, Greece
| | - Karen Angeliki Krogfelt
- Head of unit, Bacteria, Parasites & Fungi Statens Serum Institut, 5 Artillerivej, 45/112, DK-2300 Copenhagen, Denmark
| | - Konstantinos Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, BIOPOLIS, Larissa 41110, Greece
| | - Dimos Dimitrios Mitsikostas
- National and Kapodistrian University of Athens, 1st Division of Neurology, Eginition Hospital, Vasilissis Sofias 72-74, Athens 11528, Greece
| | - Georgios Hadjigeorgiou
- Department of Neurology, Faculty of Medicine, University of Thessaly, BIOPOLIS, Larissa 41110, Greece
| |
Collapse
|
50
|
The non-canonical NF-κB pathway in immunity and inflammation. NATURE REVIEWS. IMMUNOLOGY 2017. [PMID: 28580957 DOI: 10.1038/nri.2017.52)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is activated by canonical and non-canonical signalling pathways, which differ in both signalling components and biological functions. Recent studies have revealed important roles for the non-canonical NF-κB pathway in regulating different aspects of immune functions. Defects in non-canonical NF-κB signalling are associated with severe immune deficiencies, whereas dysregulated activation of this pathway contributes to the pathogenesis of various autoimmune and inflammatory diseases. Here we review the signalling mechanisms and the biological function of the non-canonical NF-κB pathway. We also discuss recent progress in elucidating the molecular mechanisms regulating non-canonical NF-κB pathway activation, which may provide new opportunities for therapeutic strategies.
Collapse
|