1
|
Rangani F, Rakhshi N, Kadkhoda Mezerji Z, Alikhah A, Dehghanzad R, Abbasi B, Ahmadi A, Nikravesh A, Pahlevan Kakhki M. Association of IL2RA and multiple sclerosis risk: A case control, systematic review, and meta-analysis study. J Neurol Sci 2025; 472:123461. [PMID: 40086235 DOI: 10.1016/j.jns.2025.123461] [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: 10/17/2024] [Revised: 02/22/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
The interleukin-2 receptor alpha chain (IL2RA) gene has been implicated in multiple sclerosis (MS) susceptibility, particularly through the rs2104286 and rs12722489 SNPs. However, previous studies have yielded inconsistent results across different populations, likely due to small sample sizes and ethnic variations. This study aimed to investigate the association of IL2RA SNPs with MS risk in eastern Iranian population and through a comprehensive meta-analysis. Our case-control study included 400 Iranian individuals from North Khorasan and Sistan & Baluchistan provinces, comprising 200 MS patients across all subtypes and genders and 200 controls. The meta-analysis incorporated pooled odds ratios (ORs) with 95 % confidence intervals (CIs) to assess the strength of these associations. Our case-control findings demonstrated a significant association between rs2104286 and MS risk, which was further supported by the meta-analysis in Iranian populations. Specifically, the association was observed at both the genotype (P = 0.001) and allelic (P = 0.003) levels in North Khorasan and at the genotype level in Sistan & Baluchistan (P = 0.001). A global meta-analysis of rs2104286, encompassing 24,931 MS patients and 36,036 controls, revealed a significant association between the A allele and all genotype models with increased MS risk (P < 0.05). Similarly, a meta-analysis of rs12722489, including 19,797 MS patients and 32,085 controls, identified the CC + CT genotype as a risk factor for MS (P = 0.04). In conclusion, our findings suggest that the rs2104286 A allele and rs12722489 CC + CT genotype are associated with an increased risk of MS in both Caucasian and Asian populations, including Iranians.
Collapse
Affiliation(s)
- Fatemeh Rangani
- Department of Clinical Neuroscience, Karolinska Institutet, and Center for Molecular Medicine, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Nahid Rakhshi
- Department of Medical Biotechnology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zahra Kadkhoda Mezerji
- Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asieh Alikhah
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine, University of Western Ontario, London, Canada
| | - Reyhaneh Dehghanzad
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Abbasi
- Department of Biology, Faculty of Basic Sciences, Zabol University, Zabol, Iran
| | - Amirhossein Ahmadi
- Department of Biological Science and Technology, Faculty of Bio and Nano Science and Technology, Persian Gulf University, Bushehr, Iran
| | - Abbas Nikravesh
- Department of Medical Biotechnology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Majid Pahlevan Kakhki
- Department of Clinical Neuroscience, Karolinska Institutet, and Center for Molecular Medicine, Karolinska University Hospital, SE-171 76 Stockholm, Sweden.
| |
Collapse
|
2
|
Eriksson AM, Emini N, Harbo HF, Berge T. Is DEXI a Multiple Sclerosis Susceptibility Gene? Int J Mol Sci 2025; 26:1175. [PMID: 39940946 PMCID: PMC11818924 DOI: 10.3390/ijms26031175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
The genetic landscape of multiple sclerosis (MS) has been extensively mapped, yielding significant insights into the molecular mechanisms of the disorder. Early studies highlighted key genes associated with the immune system, particularly T cells, as critical for MS susceptibility. Subsequent large-scale genome-wide association studies (GWASs) identified over 200 genetic variants linked to MS, revealing a complex interplay between MS risk and genes involved in various processes within adaptive and innate immune cells, as well as brain-resident microglia. Recently, a groundbreaking GWAS pinpointed the first gene variant associated with MS disease progression, distinguishing the mechanisms driving disease onset from those influencing progression. The C-type lectin domain family 16, member A (CLEC16A) gene within the 16p13 region has consistently been shown to be associated with increased risk of developing both MS and other autoimmune disorders. Notably, several autoimmune-associated genetic variants in CLEC16A introns act as expression quantitative trait loci for the dexamethasone-induced protein (DEXI gene, adding DEXI to the growing list of MS susceptibility genes. This review explores the molecular and functional characterization of DEXI with a particular focus on recent advances in understanding its role in autoimmunity, specifically in the context of multiple sclerosis. We underscore the importance of continued molecular investigation of susceptibility loci for MS identified in genetic studies, with the goal of translating this knowledge into clinical applications.
Collapse
Affiliation(s)
- Anna M. Eriksson
- Institute of Clinical Medicine, University of Oslo, Kirkeveien 166, 0450 Oslo, Norway; (A.M.E.); (H.F.H.)
- Department of Research, Østfold Hospital, Postboks 300, 1714 Grålum, Norway
| | - Nora Emini
- Department of Research, Innovation and Education, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway;
| | - Hanne F. Harbo
- Institute of Clinical Medicine, University of Oslo, Kirkeveien 166, 0450 Oslo, Norway; (A.M.E.); (H.F.H.)
- Department of Neurology, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway
| | - Tone Berge
- Department of Research, Innovation and Education, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway;
- Department of Mechanical, Electronic and Chemical Engineering, Oslo Metropolitan University, Pilestredet 35, 0166 Oslo, Norway
| |
Collapse
|
3
|
Ho MF, Zhang C, Cohan JS, Tuncturk M, Heider RM, Coombes BJ, Biernacka J, Moon I, Skime M, Ho AM, Ngo Q, Skillon C, Croarkin PE, Oesterle TS, Karpyak VM, Li H, Weinshilboum RM. IL17RB genetic variants are associated with acamprosate treatment response in patients with alcohol use disorder: A proteomics-informed genomics study. Brain Behav Immun 2024; 120:304-314. [PMID: 38852760 PMCID: PMC11269006 DOI: 10.1016/j.bbi.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Acamprosate is a Food and Drug Administration (FDA) approved medication for the treatment of alcohol use disorder (AUD). However, only a subset of patients achieves optimal treatment outcomes. Currently, no biological measures are utilized to predict response to acamprosate treatment. We applied our established pharmaco-omics informed genomics strategy to identify potential biomarkers associated with acamprosate treatment response. Specifically, our previous open-label acamprosate clinical trial recruited 442 patients with AUD who were treated with acamprosate for three months. We first performed proteomics using baseline plasma samples to identify potential biomarkers associated with acamprosate treatment outcomes. Next, we applied our established "proteomics-informed genome-wide association study (GWAS)" research strategy, and identified 12 proteins, including interleukin-17 receptor B (IL17RB), associated with acamprosate treatment response. A GWAS for IL17RB concentrations identified several genome-wide significant signals. Specifically, the top hit single nucleotide polymorphism (SNP) rs6801605 with a minor allele frequency of 38% in the European American population mapped 4 kilobase (Kb) upstream of IL17RB, and intron 1 of the choline dehydrogenase (CHDH) gene on chromosome 3 (p: 4.8E-20). The variant genotype (AA) for the SNP rs6801605 was associated with lower IL17RB protein expression. In addition, we identified a series of genetic variants in IL17RB that were associated with acamprosate treatment outcomes. Furthermore, the variantgenotypes for all of those IL17RB SNPs were protective for alcohol relapse. Finally, we demonstrated that the basal level of mRNA expression of IL17RB was inversely correlated with those of nuclear factor-κB (NF-κB) subunits, and a significantly higher expression of NF-κB subunits was observed in AUD patients who relapsed to alcohol use. In summary, this study illustrates that IL17RB genetic variants might contribute to acamprosate treatment outcomes. This series of studies represents an important step toward generating functional hypotheses that could be tested to gain insight into mechanisms underlying acamprosate treatment response phenotypes. (The ClinicalTrials.gov Identifier: NCT00662571).
Collapse
Affiliation(s)
- Ming-Fen Ho
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, USA.
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, USA
| | - James S Cohan
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Mustafa Tuncturk
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Robin M Heider
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, USA
| | - Brandon J Coombes
- Division of Computational Biology, Quantitative Health Sciences, Rochester, MN, USA
| | - Joanna Biernacka
- Division of Computational Biology, Quantitative Health Sciences, Rochester, MN, USA
| | - Irene Moon
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, USA
| | - Michelle Skime
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Ada M Ho
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Quyen Ngo
- Hazelden Betty Ford Foundation, Center City, MN, USA
| | | | - Paul E Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Tyler S Oesterle
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Victor M Karpyak
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, USA
| | | |
Collapse
|
4
|
Ma Y, Xiao Y, Zhang S, Liu J, Shang H. Association of Autoimmune Diseases with the Risk of Parkinson's Disease. Neuroepidemiology 2024:1-13. [PMID: 38981460 DOI: 10.1159/000539466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 04/29/2024] [Indexed: 07/11/2024] Open
Abstract
INTRODUCTION PD is a progressive neurodegeneration disease characterized by cardinal motor symptoms such as bradykinesia and tremor. The pathogenesis of PD remains unclear. It is hypothesized that immune system dysfunction may contribute to PD. Thus, autoimmune diseases may influence the risk of incident PD. METHODS We included 398,329 participants without PD at the baseline from UK Biobank. The association between 20 autoimmune diseases with PD was examined using cox hazards regression analyses, adjusting covariates like age, sex, and smoking status in the statistical models. Sensitivity analyses were conducted, adjusting for polygenic risk score and the reported source of PD, to check the robustness. RESULTS After an average follow-up of 13.1 ± 0.816 years, 2,245 participants were diagnosed with incident PD. After multiple comparison correction, only multiple sclerosis (MS) reached statistical significance and showed an increased risk for incident PD. Compared with non-MS patients, the risk of incident PD in MS patients was 2.57-fold with age and sex being adjusted (95% CI, 1.59-4.14; adjust p value = 0.002). After adjusting lifestyle and other factors, the hazard ratio of incident PD in MS patients was 2.49 (95% CI, 1.55-4.02; adjust p value = 0.004). Excluding the self-reported PD cases in the sensitivity analysis, MS was a detrimental factor for incident PD (HR, 2.06; 95% CI, 1.56-4.05; adjust p value = 0.004). The link between MS and PD did not reach the statistical significance in the sensitivity analysis adjusting the PRS (adjust p value = 0.95). CONCLUSION Our study provided evidence from observational analyses that MS was associated with an increased risk of PD. Further investigations should be performed to determine the causal association and potential pathophysiology between MS and PD.
Collapse
Affiliation(s)
- Yuanzheng Ma
- Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China,
- National Clinical Research Center for Geriatric, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China,
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China,
| | - Yi Xiao
- Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
- National Clinical Research Center for Geriatric, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Sirui Zhang
- Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
- National Clinical Research Center for Geriatric, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiyong Liu
- Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
- National Clinical Research Center for Geriatric, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Huifang Shang
- Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
- National Clinical Research Center for Geriatric, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Williams MJ, Orlando C, Akisanya J, Amezcua L. Multiple Sclerosis in Black and Hispanic Populations: Serving the Underserved. Neurol Clin 2024; 42:295-317. [PMID: 37980120 DOI: 10.1016/j.ncl.2023.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023]
Abstract
Multiple sclerosis has historically been characterized as a disease that affects young women of European ancestry, but recent studies indicate that the incidence and prevalence of the disease is much higher in Black and Hispanic populations than previously recognized. There is evidence that there is a more severe disease course in these populations. , but the intersection of genetic underpinnings and social determinants of health (SDOH) is poorly understood due to the lack of diversity in clinical research. Improving health disparities will involve multiple stakeholders in efforts to improve SDOH and raise awareness about research involvement and the importance of developing personalized health care plans to combat this disease.
Collapse
Affiliation(s)
- Mitzi J Williams
- Joi Life Wellness Multiple Sclerosis Center, 767 Concord Road, SE, Smyrna, GA 30082, USA.
| | - Christopher Orlando
- Department of Neurology, University of Southern California, Keck School of Medicine, 1520 San Pablo Street, Suite 3000, Los Angeles, CA, USA. https://twitter.com/OrlandoMDMPH
| | - Jemima Akisanya
- Georgetown Department of Neurology, 10401 Hospital Drive, Suite 102, Clinton, MD 20735, USA. https://twitter.com/MimasMyelin
| | - Lilyana Amezcua
- Department of Neurology, University of Southern California, Keck School of Medicine, 1520 San Pablo Street, Suite 3000, Los Angeles, CA, USA
| |
Collapse
|
6
|
Pandey R, Bakay M, Hakonarson H. SOCS-JAK-STAT inhibitors and SOCS mimetics as treatment options for autoimmune uveitis, psoriasis, lupus, and autoimmune encephalitis. Front Immunol 2023; 14:1271102. [PMID: 38022642 PMCID: PMC10643230 DOI: 10.3389/fimmu.2023.1271102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Autoimmune diseases arise from atypical immune responses that attack self-tissue epitopes, and their development is intricately connected to the disruption of the JAK-STAT signaling pathway, where SOCS proteins play crucial roles. Conditions such as autoimmune uveitis, psoriasis, lupus, and autoimmune encephalitis exhibit immune system dysfunctions associated with JAK-STAT signaling dysregulation. Emerging therapeutic strategies utilize JAK-STAT inhibitors and SOCS mimetics to modulate immune responses and alleviate autoimmune manifestations. Although more research and clinical studies are required to assess their effectiveness, safety profiles, and potential for personalized therapeutic approaches in autoimmune conditions, JAK-STAT inhibitors and SOCS mimetics show promise as potential treatment options. This review explores the action, effectiveness, safety profiles, and future prospects of JAK inhibitors and SOCS mimetics as therapeutic agents for psoriasis, autoimmune uveitis, systemic lupus erythematosus, and autoimmune encephalitis. The findings underscore the importance of investigating these targeted therapies to advance treatment options for individuals suffering from autoimmune diseases.
Collapse
Affiliation(s)
- Rahul Pandey
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Marina Bakay
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, The University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
7
|
Davis GE, Davis MJ, Lowell WE. Triggering multiple sclerosis at conception and early gestation: The variation in ultraviolet radiation is as important as its intensity. Heliyon 2023; 9:e16954. [PMID: 37346332 PMCID: PMC10279836 DOI: 10.1016/j.heliyon.2023.e16954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 04/17/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023] Open
Abstract
Background and objectives Medical science needs to further elucidate the role of ultraviolet radiation (UVR), geographic latitude, and the role of vitamin D in the autoimmune disease multiple sclerosis (MS). We separated several papers into categories out of the thousands published and used their conclusions to explore the relationship between UVR and MS. Relevance MS is increasing in incidence, particularly in women where MS is two to three times that in men and particularly severe in African Americans. Methods We collected UVR data at our observatory in Central Maine and calculated the average coefficient of variation (CVUVR) for each month for 15 years (2007-2021, inclusive). Results The month of conception (MOC) is more important than the month of birth (MOB) in explaining how UVR triggers the variable genetic predisposition to MS. We hypothesize that the rapidly increasing CVUVR is important in preventing an increase in the activity of the vitamin D receptor (VDR) from August to December, which then requires a higher intensity of UVR later in life to suppress the immune system, therefore predisposing to more MS. Limitations One observatory at about 44° latitude. Conclusions While variation in UVR is important at the MOC if UVR exceeds a threshold (e.g., if the sunspot number equals or is greater than 90, usually at a solar cycle MAX, or at elevations above approximately 3,000 feet above sea level), the MS mitigating vitamin D-VDR mechanism is overwhelmed and the genotoxic effects of higher-intensity UVR promote MS in those with a genetic predisposition. What is new in this research This paper offers a new concept in MS research.
Collapse
Affiliation(s)
- George E. Davis
- Riverview Psychiatric Center, 250 Arsenal Street, State House Station #11, Augusta, ME, 04333-0011, USA
| | - Matthew J. Davis
- Riverview Psychiatric Center, 250 Arsenal Street, State House Station #11, Augusta, ME, 04333-0011, USA
| | - Walter E. Lowell
- Riverview Psychiatric Center, 250 Arsenal Street, State House Station #11, Augusta, ME, 04333-0011, USA
| |
Collapse
|
8
|
Telesford KM, Amezcua L, Tardo L, Horton L, Lund BT, Reder AT, Vartanian T, Monson NL. Understanding humoral immunity and multiple sclerosis severity in Black, and Latinx patients. Front Immunol 2023; 14:1172993. [PMID: 37215103 PMCID: PMC10196635 DOI: 10.3389/fimmu.2023.1172993] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
People identified with Black/African American or Hispanic/Latinx ethnicity are more likely to exhibit a more severe multiple sclerosis disease course relative to those who identify as White. While social determinants of health account for some of this discordant severity, investigation into contributing immunobiology remains sparse. The limited immunologic data stands in stark contrast to the volume of clinical studies describing ethnicity-associated discordant presentation, and to advancement made in our understanding of MS immunopathogenesis over the past several decades. In this perspective, we posit that humoral immune responses offer a promising avenue to better understand underpinnings of discordant MS severity among Black/African American, and Hispanic/Latinx-identifying patients.
Collapse
Affiliation(s)
- Kiel M. Telesford
- Department of Neurology, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Lilyana Amezcua
- Multiple Sclerosis Comprehensive Care Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lauren Tardo
- Department of Neurology, University of Texas Southwestern Medical Center (UT), Dallas, TX, United States
| | - Lindsay Horton
- Department of Neurology, University of Texas Southwestern Medical Center (UT), Dallas, TX, United States
| | - Brett T. Lund
- Multiple Sclerosis Comprehensive Care Center, University of Southern California, Los Angeles, CA, United States
| | - Anthony T. Reder
- Department of Neurology, University of Chicago, Chicago, IL, United States
| | - Timothy Vartanian
- Department of Neurology, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Nancy L. Monson
- Department of Neurology, University of Texas Southwestern Medical Center (UT), Dallas, TX, United States
| |
Collapse
|
9
|
Pandey R, Bakay M, Hakonarson H. CLEC16A-An Emerging Master Regulator of Autoimmunity and Neurodegeneration. Int J Mol Sci 2023; 24:ijms24098224. [PMID: 37175930 PMCID: PMC10179542 DOI: 10.3390/ijms24098224] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
CLEC16A is emerging as an important genetic risk factor for several autoimmune disorders and for Parkinson disease (PD), opening new avenues for translational research and therapeutic development. While the exact role of CLEC16A in health and disease is still being elucidated, the gene plays a critical role in the regulation of autophagy, mitophagy, endocytosis, intracellular trafficking, immune function, and in biological processes such as insulin secretion and others that are important to cellular homeostasis. As shown in both human and animal modeling studies, CLEC16A hypofunction predisposes to both autoinflammatory phenotype and neurodegeneration. While the two are clearly related, further functional studies are needed to fully understand the mechanisms involved for optimized therapeutic interventions. Based on recent data, mitophagy-inducing drugs may be warranted, and such therapy should be tested in clinical trials as these drugs would tackle the underlying pathogenic mechanism (s) and could treat or prevent symptoms of autoimmunity and neurodegeneration in individuals with CLEC16A risk variants. Accordingly, interventions directed at reversing the dysregulated mitophagy and the consequences of loss of function of CLEC16A without activating other detrimental cellular pathways could present an effective therapy. This review presents the emerging role of CLEC16A in health and disease and provides an update on the disease processes that are attributed to variants located in the CLEC16A gene, which are responsible for autoimmune disorders and neurodegeneration with emphasis on how this information is being translated into practical and effective applications in the clinic.
Collapse
Affiliation(s)
- Rahul Pandey
- Center for Applied Genomics, Children's Hospital of Philadelphia, Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104-4318, USA
| | - Marina Bakay
- Center for Applied Genomics, Children's Hospital of Philadelphia, Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104-4318, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104-4318, USA
- Department of Pediatrics, The University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4318, USA
| |
Collapse
|
10
|
Xiao H, Wang G, Zhao M, Shuai W, Ouyang L, Sun Q. Ras superfamily GTPase activating proteins in cancer: Potential therapeutic targets? Eur J Med Chem 2023; 248:115104. [PMID: 36641861 DOI: 10.1016/j.ejmech.2023.115104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
To search more therapeutic strategies for Ras-mutant tumors, regulators of the Ras superfamily involved in the GTP/GDP (guanosine triphosphate/guanosine diphosphate) cycle have been well concerned for their anti-tumor potentials. GTPase activating proteins (GAPs) provide the catalytic group necessary for the hydrolysis of GTPs, which accelerate the switch by cycling between GTP-bound active and GDP-bound inactive forms. Inactivated GAPs lose their function in activating GTPase, leading to the continuous activation of downstream signaling pathways, uncontrolled cell proliferation, and eventually carcinogenesis. A growing number of evidence has shown the close link between GAPs and human tumors, and as a result, GAPs are believed as potential anti-tumor targets. The present review mainly summarizes the critically important role of GAPs in human tumors by introducing the classification, function and regulatory mechanism. Moreover, we comprehensively describe the relationship between dysregulated GAPs and the certain type of tumor. Finally, the current status, research progress, and clinical value of GAPs as therapeutic targets are also discussed, as well as the challenges and future direction in the cancer therapy.
Collapse
Affiliation(s)
- Huan Xiao
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Min Zhao
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Wen Shuai
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
11
|
von Werdt D, Gungor B, Barreto de Albuquerque J, Gruber T, Zysset D, Kwong Chung CKC, Corrêa-Ferreira A, Berchtold R, Page N, Schenk M, Kehrl JH, Merkler D, Imhof BA, Stein JV, Abe J, Turchinovich G, Finke D, Hayday AC, Corazza N, Mueller C. Regulator of G-protein signaling 1 critically supports CD8 + T RM cell-mediated intestinal immunity. Front Immunol 2023; 14:1085895. [PMID: 37153600 PMCID: PMC10158727 DOI: 10.3389/fimmu.2023.1085895] [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: 10/31/2022] [Accepted: 02/13/2023] [Indexed: 05/09/2023] Open
Abstract
Members of the Regulator of G-protein signaling (Rgs) family regulate the extent and timing of G protein signaling by increasing the GTPase activity of Gα protein subunits. The Rgs family member Rgs1 is one of the most up-regulated genes in tissue-resident memory (TRM) T cells when compared to their circulating T cell counterparts. Functionally, Rgs1 preferentially deactivates Gαq, and Gαi protein subunits and can therefore also attenuate chemokine receptor-mediated immune cell trafficking. The impact of Rgs1 expression on tissue-resident T cell generation, their maintenance, and the immunosurveillance of barrier tissues, however, is only incompletely understood. Here we report that Rgs1 expression is readily induced in naïve OT-I T cells in vivo following intestinal infection with Listeria monocytogenes-OVA. In bone marrow chimeras, Rgs1 -/- and Rgs1 +/+ T cells were generally present in comparable frequencies in distinct T cell subsets of the intestinal mucosa, mesenteric lymph nodes, and spleen. After intestinal infection with Listeria monocytogenes-OVA, however, OT-I Rgs1 +/+ T cells outnumbered the co-transferred OT-I Rgs1- /- T cells in the small intestinal mucosa already early after infection. The underrepresentation of the OT-I Rgs1 -/- T cells persisted to become even more pronounced during the memory phase (d30 post-infection). Remarkably, upon intestinal reinfection, mice with intestinal OT-I Rgs1 +/+ TRM cells were able to prevent the systemic dissemination of the pathogen more efficiently than those with OT-I Rgs1 -/- TRM cells. While the underlying mechanisms are not fully elucidated yet, these data thus identify Rgs1 as a critical regulator for the generation and maintenance of tissue-resident CD8+ T cells as a prerequisite for efficient local immunosurveillance in barrier tissues in case of reinfections with potential pathogens.
Collapse
Affiliation(s)
- Diego von Werdt
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Bilgi Gungor
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Thomas Gruber
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Daniel Zysset
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Cheong K. C. Kwong Chung
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Gastrointestinal Health, Immunology, Nestlé Research, Lausanne, Switzerland
| | - Antonia Corrêa-Ferreira
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Regina Berchtold
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Nicolas Page
- Department of Pathology, Division of Clinical Pathology, University & University Hospitals of Geneva, Geneva, Switzerland
| | - Mirjam Schenk
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - John H. Kehrl
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Doron Merkler
- Department of Pathology, Division of Clinical Pathology, University & University Hospitals of Geneva, Geneva, Switzerland
| | - Beat A. Imhof
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Pathology and Immunology, Centre Medical Universitaire, University of Geneva, Geneva, Switzerland
| | - Jens V. Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Gleb Turchinovich
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniela Finke
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
| | - Adrian C. Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | - Nadia Corazza
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- *Correspondence: Christoph Mueller, ; Nadia Corazza,
| | - Christoph Mueller
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
- *Correspondence: Christoph Mueller, ; Nadia Corazza,
| |
Collapse
|
12
|
Timasheva YR, Nasibullin TR, Tuktarova IA, Erdman VV, Galiullin TR, Lyutov OV, Bakhtiiarova KZ. [Multiple sclerosis in the Republic of Bashkortostan: population-specific genetic predictors and the results of a 20-year clinical follow-up study]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:34-42. [PMID: 37560832 DOI: 10.17116/jnevro202312307234] [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] [Indexed: 08/11/2023]
Abstract
OBJECTIVE Identification of a complex of genetic predictors of multiple sclerosis (MS) based on previously obtained results in genome-wide association studies of disease markers (GWAS markers) in a population of MS patients and healthy individuals of the Republic of Bashkortostan (Russian Federation) using polygenic detection. MATERIAL AND METHODS The total study group consisted of 2048 people (641 patients with MS and 1407 healthy individuals) who permanently resided in the Republic of Bashkortostan and belonged to the Bashkir (n=325), Russian (n=772) or Tatar (n=951) nationalities. The analysis of association between MS and polymorphisms previously associated with the disease according to GWAS data was performed. Of the 641 MS patients, 247 were the subject of a 20-year prospective clinical follow-up. RESULTS The C6orf10 rs3129934*T allele was most significantly associated with MS in Russians (OR=2.00, P=5.85·10-5) and Tatars (OR=2.38, P=8.61·10-7). An increased MS risk in Russians was also associated with the EOMES rs11129295*T (OR=1.56, P=0.007) and IL7R rs1494558*I (OR=1.61, P=0.003) alleles. Meta-analysis confirmed the association of the C6orf10 rs3129934*T, EOMES rs11129295*T and IL7R rs1494558*I alleles with MS in the total group, as well as revealed associations of the INAVA rs7522462*G, IL7R rs10624573*I, CD6 rs17824933*G, GPC5 rs9523762*A and GPR65 rs2119704*C alleles with the disease. Using polygenic analysis, we identified a complex predictor C6orf10 rs3129934*C + INAVA rs7522462*G + CD6 rs17824933*C with a pronounced protective effect against MS in the total group (OR=0.34, PFDR=2.65·10-7). CONCLUSION We reproduced the association of eight polymorphisms (C6orf10 rs3129934, INAVA rs7522462, IL7R rs10624573, EOMES rs11129295, GPR65 rs2119704, GPC5 rs9523762, CD6 rs17824933 and CD58 rs2300747) with MS, previously identified in GWAS in European populations. Whole exome or genome sequencing may help to reveal the mechanisms underlying the pathogenesis of MS in populations of the Russian Federation.
Collapse
Affiliation(s)
- Y R Timasheva
- Institute of Biochemistry and Genetics of Ufa Federal Research Centre, Ufa, Russia
- Bashkir State Medical University, Ufa, Russia
| | - T R Nasibullin
- Institute of Biochemistry and Genetics of Ufa Federal Research Centre, Ufa, Russia
| | - I A Tuktarova
- Institute of Biochemistry and Genetics of Ufa Federal Research Centre, Ufa, Russia
| | - V V Erdman
- Institute of Biochemistry and Genetics of Ufa Federal Research Centre, Ufa, Russia
| | | | - O V Lyutov
- Bashkir State Medical University, Ufa, Russia
| | | |
Collapse
|
13
|
Das S, Taylor K, Kozubek J, Sardell J, Gardner S. Genetic risk factors for ME/CFS identified using combinatorial analysis. J Transl Med 2022; 20:598. [PMID: 36517845 PMCID: PMC9749644 DOI: 10.1186/s12967-022-03815-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic disease that lacks known pathogenesis, distinctive diagnostic criteria, and effective treatment options. Understanding the genetic (and other) risk factors associated with the disease would begin to help to alleviate some of these issues for patients. METHODS We applied both GWAS and the PrecisionLife combinatorial analytics platform to analyze ME/CFS cohorts from UK Biobank, including the Pain Questionnaire cohort, in a case-control design with 1000 cycles of fully random permutation. Results from this study were supported by a series of replication and cohort comparison experiments, including use of disjoint Verbal Interview CFS, post-viral fatigue syndrome and fibromyalgia cohorts also derived from UK Biobank, and compared results for overlap and reproducibility. RESULTS Combinatorial analysis revealed 199 SNPs mapping to 14 genes that were significantly associated with 91% of the cases in the ME/CFS population. These SNPs were found to stratify by shared cases into 15 clusters (communities) made up of 84 high-order combinations of between 3 and 5 SNPs. p-values for these communities range from 2.3 × 10-10 to 1.6 × 10-72. Many of the genes identified are linked to the key cellular mechanisms hypothesized to underpin ME/CFS, including vulnerabilities to stress and/or infection, mitochondrial dysfunction, sleep disturbance and autoimmune development. We identified 3 of the critical SNPs replicated in the post-viral fatigue syndrome cohort and 2 SNPs replicated in the fibromyalgia cohort. We also noted similarities with genes associated with multiple sclerosis and long COVID, which share some symptoms and potentially a viral infection trigger with ME/CFS. CONCLUSIONS This study provides the first detailed genetic insights into the pathophysiological mechanisms underpinning ME/CFS and offers new approaches for better diagnosis and treatment of patients.
Collapse
Affiliation(s)
- Sayoni Das
- PrecisionLife Ltd, Long Hanborough, Oxford, UK
| | | | | | | | | |
Collapse
|
14
|
Jacobs BM, Peter M, Giovannoni G, Noyce AJ, Morris HR, Dobson R. Towards a global view of multiple sclerosis genetics. Nat Rev Neurol 2022; 18:613-623. [PMID: 36075979 DOI: 10.1038/s41582-022-00704-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a neuroimmunological disorder of the CNS with a strong heritable component. The genetic architecture of MS susceptibility is well understood in populations of European ancestry. However, the extent to which this architecture explains MS susceptibility in populations of non-European ancestry remains unclear. In this Perspective article, we outline the scientific arguments for studying MS genetics in ancestrally diverse populations. We argue that this approach is likely to yield insights that could benefit individuals with MS from all ancestral groups. We explore the logistical and theoretical challenges that have held back this field to date and conclude that, despite these challenges, inclusion of participants of non-European ancestry in MS genetics studies will ultimately be of value to all patients with MS worldwide.
Collapse
Affiliation(s)
- Benjamin Meir Jacobs
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University London, London, UK. .,Department of Neurology, Royal London Hospital, London, UK.
| | - Michelle Peter
- NHS North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Gavin Giovannoni
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University London, London, UK.,Department of Neurology, Royal London Hospital, London, UK.,Blizard Institute, Queen Mary University London, London, UK
| | - Alastair J Noyce
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University London, London, UK.,Department of Neurology, Royal London Hospital, London, UK.,Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Huw R Morris
- Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Ruth Dobson
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University London, London, UK.,Department of Neurology, Royal London Hospital, London, UK
| |
Collapse
|
15
|
Casadó-Llombart S, Gheitasi H, Ariño S, Consuegra-Fernández M, Armiger-Borràs N, Kostov B, Ramos-Casals M, Brito-Zerón P, Lozano F. Gene Variation at Immunomodulatory and Cell Adhesion Molecules Loci Impacts Primary Sjögren's Syndrome. Front Med (Lausanne) 2022; 9:822290. [PMID: 35372412 PMCID: PMC8971656 DOI: 10.3389/fmed.2022.822290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/16/2022] [Indexed: 11/24/2022] Open
Abstract
Primary Sjögren's syndrome (pSS) is an autoimmune disease triggered by a combination of environmental and host genetic factors, which results in the focal lymphocytic infiltration of exocrine glands causing eye and mouth dryness. Glandular infiltrates include T and B cell subsets positive for CD5 and/or CD6, two surface scavenger receptors involved in the fine-tuning of intracellular signals mediated by the antigen-specific receptor complex of T (TCR) and B (BCR) cells. Moreover, the epithelial cells of inflamed glands overexpress CD166/ALCAM, a CD6 ligand involved in homo and heterotypic cell adhesion interactions. All this, together with the reported association of functionally relevant single nucleotide polymorphisms (SNPs) of CD5, CD6, and CD166/ALCAM with the risk or prognosis of some immune-mediated inflammatory disorders, led us to investigate similar associations in a local cohort of patients with pSS. The logistic regression analyses of individual SNPs showed the association of CD5 rs2241002T with anti-Ro/La positivity, CD6 rs17824933C with neutropenia, and CD6 rs11230563T with increased leukopenia and neutropenia but decreased peripheral nervous system EULAR Sjögren's syndrome disease activity index (ESSDAI). Further analyses showed the association of haplotypes from CD5 (rs2241002T-rs2229177C) with anemia and thrombocytopenia, CD6 (rs17824933G-rs11230563C-rs12360861G) with cutaneous ESSDAI, and CD166/ALCAM (rs6437585C-rs579565A-rs1044243C and rs6437585C-rs579565G-rs1044243T) with disease susceptibility and several analytical parameters (anti-nuclear antibodies, neurological ESSDAI, and hematologic cytopenias). These results support the relevance of gene variation at loci coding for cell surface receptors involved in the modulation of T and B lymphocyte activation (CD5, CD6) and epithelial-immune cell adhesion (CD166/ALCAM) in modulating the clinical and analytical outcomes in patients with pSS.
Collapse
Affiliation(s)
- Sergi Casadó-Llombart
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Hoda Gheitasi
- Department of Autoimmune Diseases, ICMiD, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Silvia Ariño
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marta Consuegra-Fernández
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Noelia Armiger-Borràs
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Belchin Kostov
- Primary Care Centre Les Corts, Consorci d'Atenció Primària de Salut Barcelona Esquerra (CAPSBE), Barcelona, Spain
- Primary Healthcare Transversal Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Statistics and Operations Research, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
| | - Manuel Ramos-Casals
- Department of Autoimmune Diseases, ICMiD, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Pilar Brito-Zerón
- Research and Innovation Group in Autoimmune Diseases, RGAD-Sanitas Digital Hospital, Barcelona, Spain
- Systemic Autoimmune Diseases Unit, Internal Medicine, Millenium Clinic, Sanitas, Barcelona, Spain
- *Correspondence: Pilar Brito-Zerón
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Servei d'Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Francisco Lozano
| |
Collapse
|
16
|
Borrelia burgdorferi is a poor inducer of interferon-gamma: amplification induced by interleukin-12. Infect Immun 2022; 90:e0055821. [PMID: 35130450 DOI: 10.1128/iai.00558-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background Laboratory diagnosis of Lyme borreliosis (LB) is mainly based on serology, which has limitations, particularly in the early stages of the disease. In recent years there have been conflicting reports concerning a new diagnostic tool using the cytokine interferon-gamma (IFN-γ). Previous studies have generally found low concentrations of IFN-γ in early LB infection. The goal of this study is to investigate IFN-γ regulation during early LB and provide insights into the host response to B. burgdorferi. Methods We performed in vitro experiments with whole blood assays and peripheral blood mononuclear cells (PBMCs) of LB patients and healthy volunteers exposed to B. burgdorferi and evaluated the IFN-γ response using ELISA and related interindividual variation in IFN-γ production to the presence of single nucleotide polymorphisms. Results IFN-γ production of B. burgdorferi-exposed PBMCs and whole blood was amplified by the addition of IL-12 to the stimulation system. This effect was observed after 24 hours of B. burgdorferi stimulation in both healthy individuals and LB patients. The effect was highly variable between individuals, but was significantly higher in LB patients six weeks since the start of antibiotic treatment compared to healthy individuals. IL-12 p40 and IL-18 mRNA was upregulated upon exposure to B. burgdorferi, whereas IL-12 p35 and IFN-γ mRNA expression remained relatively unchanged. SNP Rs280520 in the downstream IL-12 pathway, Tyrosine Kinase 2, was associated with increased IFN-γ production. Conclusions This study shows that IL-12 evokes an IFN-γ response in B. burgdorferi exposed cells, and LB patients and healthy controls respond differently to this stimulation.
Collapse
|
17
|
Chase C, Connell E, Elliott SN, Jones LK, Larinde O, Musachia AM, Smith EA, Cofield SS, Wingo BC. Differences in Cardiometabolic Comorbidities Between Black and White Persons Living With Multiple Sclerosis. Arch Phys Med Rehabil 2022; 103:331-335. [PMID: 34728190 PMCID: PMC9808816 DOI: 10.1016/j.apmr.2021.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/10/2021] [Accepted: 10/04/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To determine differences in obesity, type 2 diabetes, and hypertension in Black patients compared with White patients with multiple sclerosis (MS). DESIGN Cross-sectional database review. SETTING Large academic medical center research records database. PARTICIPANTS A total of 3191 patient cases (N=3191; 77% female, 34% Black) identified by MS diagnosis within the medical record. INTERVENTIONS Not applicable. MAIN OUTCOME MEASURES Diagnosis codes for type 2 diabetes and hypertension. Body mass index (BMI), race, age, and sex were collected. Analysis of variance (continuous variables) and chi-square analyses (categorical variables) were conducted to determine differences in obesity, diabetes, and hypertension between race and sex. Logistic regression was conducted to determine odds ratios (ORs) of developing diabetes and hypertension based on race, sex, BMI, and age. RESULTS Black patients were more than twice as likely to be diagnosed as having diabetes (OR, 2.15 [95% CI, 1.70-2.72]; P<.0001) or hypertension (OR, 2.44 [95% CI, 2.05-2.91], P<.0001) compared with White patients. Sex did not present a greater likelihood of being diagnosed as having diabetes; however, men were 1.22 times more likely be diagnosed as having hypertension compared with women (95% CI, 1.01-1.49; P=.0439). Increased age and BMI were also significantly associated with likelihood of diagnosis of diabetes and hypertension (age: diabetes OR, 1.05 [95% CI, 1.04-1.06], P<.0001; hypertension OR, 1.06 [95% CI, 1.05-1.06], P<.0001; BMI: diabetes obese vs normal: OR, 2.11 [95% CI, 1.43-3.11], P=.0002; hypertension: obese vs normal: OR, 1.72 [95% CI, 1.39-2.13], P<.0001). CONCLUSIONS Black patients with MS are significantly more likely to have cardiometabolic conditions than White patients. These conditions have been associated with poorer health outcomes for people with MS and may have some effect on the differences in MS disease course reported in Black patients.
Collapse
Affiliation(s)
- Catherine Chase
- Department of Occupational Therapy, School of Health Professions, University of Alabama at Birmingham, Birmingham, Alabama
| | - Erika Connell
- Department of Occupational Therapy, School of Health Professions, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sabrina N Elliott
- Department of Occupational Therapy, School of Health Professions, University of Alabama at Birmingham, Birmingham, Alabama
| | - Laura-Katherine Jones
- Department of Occupational Therapy, School of Health Professions, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Ashley M Musachia
- Department of Occupational Therapy, School of Health Professions, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elizabeth Ann Smith
- Department of Occupational Therapy, School of Health Professions, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stacey S Cofield
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham
| | - Brooks C Wingo
- Department of Occupational Therapy, School of Health Professions, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
18
|
Garrick O, Mesa R, Ferris A, Kim ES, Mitchell E, Brawley OW, Carpten J, Carter KD, Coney J, Winn R, Monroe S, Sandoval F, Perez E, Williams M, Grove E, Highsmith Q, Richie N, Begelman SM, Collins AS, Freedman J, Gonzales MS, Wilson G. Advancing Inclusive Research: Establishing Collaborative Strategies to Improve Diversity in Clinical Trials. Ethn Dis 2022; 32:61-68. [PMID: 35106045 PMCID: PMC8785867 DOI: 10.18865/ed.32.1.61] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023] Open
Abstract
Well-characterized disparities in clinical research have disproportionately affected patients of color, particularly in underserved communities. To tackle these barriers, Genentech formed the External Council for Advancing Inclusive Research, a 14-person committee dedicated to developing strategies to increase clinical research participation. To help improve the recruitment and retention of patients of color, this article chronicles our efforts to tangibly address the clinical research barriers at the system, study, and patient levels over the last four years. These efforts are one of the initial steps to fully realize the promise of personalized health care and provide increased patient benefit at less cost to society. Instead of simply acknowledging the problem, here we illuminate the collaborative and multilevel strategies that have been effective in delivering meaningful progress for patients.
Collapse
Affiliation(s)
| | - Ruben Mesa
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX
| | | | | | - Edith Mitchell
- Sidney Kimmel Cancer Center – Jefferson Health, Philadelphia, PA
| | - Otis W. Brawley
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - John Carpten
- Department of Translational Genomics, Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Keith D. Carter
- Department of Ophthalmology, University of Iowa, Iowa City, IA
| | | | - Robert Winn
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA
| | | | | | - Edith Perez
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Genetics and functional genomics of multiple sclerosis. Semin Immunopathol 2022; 44:63-79. [PMID: 35022889 DOI: 10.1007/s00281-021-00907-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory neurodegenerative disease with genetic predisposition. Over the last decade, genome-wide association studies with increasing sample size led to the discovery of robustly associated genetic variants at an exponential rate. More than 200 genetic loci have been associated with MS susceptibility and almost half of its heritability can be accounted for. However, many challenges and unknowns remain. Definitive studies of disease progression and endophenotypes are yet to be performed, whereas the majority of the identified MS variants are not yet functionally characterized. Despite these shortcomings, the unraveling of MS genetics has opened up a new chapter on our understanding MS causal mechanisms.
Collapse
|
20
|
Eriksson AM, Leikfoss IS, Abrahamsen G, Sundvold V, Isom MM, Keshari PK, Rognes T, Landsverk OJB, Bos SD, Harbo HF, Spurkland A, Berge T. Exploring the role of the multiple sclerosis susceptibility gene CLEC16A in T cells. Scand J Immunol 2021; 94:e13050. [PMID: 34643957 DOI: 10.1111/sji.13050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/29/2022]
Abstract
C-type lectin-like domain family 16 member A (CLEC16A) is associated with autoimmune disorders, including multiple sclerosis (MS), but its functional relevance is not completely understood. CLEC16A is expressed in several immune cells, where it affects autophagic processes and receptor expression. Recently, we reported that the risk genotype of an MS-associated single nucleotide polymorphism in CLEC16A intron 19 is associated with higher expression of CLEC16A in CD4+ T cells. Here, we show that CLEC16A expression is induced in CD4+ T cells upon T cell activation. By the use of imaging flow cytometry and confocal microscopy, we demonstrate that CLEC16A is located in Rab4a-positive recycling endosomes in Jurkat TAg T cells. CLEC16A knock-down in Jurkat cells resulted in lower cell surface expression of the T cell receptor, however, this did not have a major impact on T cell activation response in vitro in Jurkat nor in human, primary CD4+ T cells.
Collapse
Affiliation(s)
- Anna M Eriksson
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingvild Sørum Leikfoss
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Neuroscience Research Unit, Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway
| | - Greger Abrahamsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Vibeke Sundvold
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Pankaj K Keshari
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Rognes
- Department of Informatics, University of Oslo, Oslo, Norway.,Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | | | - Steffan D Bos
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hanne F Harbo
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Spurkland
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Tone Berge
- Neuroscience Research Unit, Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway.,Department of Mechanical, Electronic and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
21
|
Liver kinase B1 rs9282860 polymorphism and risk for multiple sclerosis in White and Black Americans. Mult Scler Relat Disord 2021; 55:103185. [PMID: 34371271 DOI: 10.1016/j.msard.2021.103185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/12/2021] [Accepted: 07/31/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND We previously reported that the single nucleotide polymorphism (SNP) rs9282860 in serine threonine kinase 11 (STK11) gene which codes for liver kinase B1 (LKB1) has higher prevalence in White relapsing-remitting multiple sclerosis (RRMS) patients than controls. However it is not known if this SNP is a risk factor for MS in other populations. METHODS We assessed the prevalence of the STK11 SNP in samples collected from African American (AA) persons with MS (PwMS) and controls at multiple Veterans Affairs (VA) Medical Centers and from a network of academic MS centers. Genotyping was carried out using a specific Taqman assay. Comparisons of SNP frequencies were made using Fisher's exact test to determine significance and odds ratios. Group means were compared by appropriate t-tests based on normality and variance using SPSS V27. RESULTS There were no significant differences in average age at first symptom onset, age at diagnosis, disease duration, or disease severity between RRMS patients recruited from VAMCs versus non-VAMCs. The SNP was more prevalent in AA than White PwMS, however only in secondary progressive MS (SPMS) patients was that difference statistically significant. AA SPMS patients had higher STK11 SNP prevalence than controls; and in that cohort the SNP was associated with older age at symptom onset and at diagnosis. CONCLUSIONS The results suggest that the STK11 SNP represents a risk factor for SPMS in AA patients, and can influence both early (onset) and later (conversion to SPMSS) events.
Collapse
|
22
|
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
|
23
|
Contribution of Evolutionary Selected Immune Gene Polymorphism to Immune-Related Disorders: The Case of Lymphocyte Scavenger Receptors CD5 and CD6. Int J Mol Sci 2021; 22:ijms22105315. [PMID: 34070159 PMCID: PMC8158487 DOI: 10.3390/ijms22105315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 01/23/2023] Open
Abstract
Pathogens are one of the main selective pressures that ancestral humans had to adapt to. Components of the immune response system have been preferential targets of natural selection in response to such pathogen-driven pressure. In turn, there is compelling evidence showing that positively selected immune gene variants conferring increased resistance to past or present infectious agents are today associated with increased risk for autoimmune or inflammatory disorders but decreased risk of cancer, the other side of the same coin. CD5 and CD6 are lymphocytic scavenger receptors at the interphase of the innate and adaptive immune responses since they are involved in both: (i) microbial-associated pattern recognition; and (ii) modulation of intracellular signals mediated by the clonotypic antigen-specific receptor present in T and B cells (TCR and BCR, respectively). Here, we review available information on CD5 and CD6 as targets of natural selection as well as on the role of CD5 and CD6 variation in autoimmunity and cancer.
Collapse
|
24
|
Cree BAC, Pradhan A, Pei J, Williams MJ. Efficacy and safety of ocrelizumab vs interferon beta-1a in participants of African descent with relapsing multiple sclerosis in the Phase III OPERA I and OPERA II studies. Mult Scler Relat Disord 2021; 52:103010. [PMID: 34147885 DOI: 10.1016/j.msard.2021.103010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/05/2021] [Accepted: 04/30/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND People of African descent with multiple sclerosis (MS) appear to have a more severe disease course and may have an attenuated response to some medications compared with people of European descent. METHODS This is a post hoc subgroup analysis of participants of African descent with relapsing forms of MS who were enrolled in the Phase III OPERA I or OPERA II clinical trials and treated with ocrelizumab (OCR) 600 mg every 6 months or interferon beta-1a (IFN β-1a) 44 μg 3 times per week. RESULTS Among the 1,656 participants enrolled in OPERA I and II, 72 (4.3%) were of African descent (OCR, 40; IFN β-1a, 32). A trend for reduction in annualized relapse rate (ARR) was observed in participants of African descent, with an ≈50% reduction with OCR vs IFN β-1a. The relative rate of the mean number of gadolinium-enhancing lesions on magnetic resonance imaging (MRI) was 0.04 (95% CI, 0.01-0.22; p=0.001) in participants of African descent treated with OCR compared with IFN β-1a. Similarly, the relative rate of the number of new or enlarging T2 lesions on MRI was 0.14 (95% CI, 0.06-0.32; p<0.001). In participants of African descent, those treated with OCR were 2.61 times more likely than those who received IFN β-1a to be classified as having no evidence of disease activity (95% CI, 1.24-5.49; p=0.003) and 4.17 times more likely to be classified as having no evidence of disease activity or progression (95% CI, 1.27-13.65; p=0.006). African-descent participants tended to have a greater radiographic burden of disease at baseline, develop more brain lesions when treated with IFN β-1a, and be at greater risk of disability progression than non-African-descent participants. Participants of African descent experienced slightly more adverse events, serious adverse events, and hypersensitivity reactions than non-African-descent participants. CONCLUSION In this small sample of participants of African descent with relapsing MS from the OPERA studies, OCR demonstrated treatment benefits in clinical, MRI, and composite efficacy outcomes vs IFN β-1a, consistent with what was observed in the complete OPERA intention-to-treat cohorts.
Collapse
Affiliation(s)
- Bruce A C Cree
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA.
| | | | - Jinglan Pei
- Genentech, Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
25
|
Pellenz FM, Dieter C, Lemos NE, Bauer AC, Souza BMD, Crispim D. Association of TYK2 polymorphisms with autoimmune diseases: A comprehensive and updated systematic review with meta-analysis. Genet Mol Biol 2021; 44:e20200425. [PMID: 33949620 PMCID: PMC8097517 DOI: 10.1590/1678-4685-gmb-2020-0425] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/09/2021] [Indexed: 12/05/2022] Open
Abstract
Autoimmune diseases are characterized by the loss of self-tolerance, leading to
immune-mediated tissue destruction and chronic inflammation. Tyrosine kinase 2
(TYK2) protein plays a key role in immunity and apoptosis pathways. Studies have
reported associations between single nucleotide polymorphisms (SNPs) in the
TYK2 gene and autoimmune diseases; however, results are
still inconclusive. Thus, we conducted a systematic review followed by
meta-analysis. A literature search was performed to find studies that
investigated associations between TYK2 SNPs and autoimmune
diseases (multiple sclerosis, systemic lupus erythematosus, Crohn’s disease,
ulcerative colitis, psoriasis, rheumatoid arthritis, type 1 diabetes, and
inflammatory bowel disease). Pooled odds ratios (OR) with 95 % CI were
calculated using random (REM) or fixed (FEM) effects models in the Stata 11.0
Software. Thirty-four articles were eligible for inclusion in the meta-analyses,
comprising 9 different SNPs: rs280496, rs280500, rs280523, rs280519, rs2304256,
rs12720270, rs12720356, rs34536443, and rs35018800. Meta-analysis results showed
the minor alleles of rs2304256, rs12720270, rs12720356, rs34536443, and
rs35018800 SNPs were associated with protection against autoimmune diseases.
Moreover, the A allele of the rs280519 SNP was associated with risk for systemic
lupus erythematosus. Our meta-analyses demonstrated that the rs2304256,
rs12720270, rs12720356, rs34536443, rs35018800, and rs280519 SNPs in the
TYK2 gene are associated with different autoimmune
diseases.
Collapse
Affiliation(s)
- Felipe Mateus Pellenz
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Cristine Dieter
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Natália Emerim Lemos
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Andrea Carla Bauer
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil.,Hospital de Clínicas de Porto Alegre, Serviço de Nefrologia, Porto Alegre, RS, Brazil
| | - Bianca Marmontel de Souza
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Daisy Crispim
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| |
Collapse
|
26
|
Correlation between contrast enhanced plaques and plaque diffusion restriction and their signal intensities in FLAIR images in patients who admitted with acute symptoms of multiple sclerosis. J Med Imaging Radiat Sci 2021; 52:121-126. [DOI: 10.1016/j.jmir.2020.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 10/26/2020] [Accepted: 12/15/2020] [Indexed: 11/24/2022]
|
27
|
Peng Y, Chen B, Sheng X, Qian Y. Polymorphisms in IRF5 and TYK2 Genes Are Associated with Rheumatoid Arthritis in a Chinese Han Population. Med Sci Monit 2021; 27:e928455. [PMID: 33583939 PMCID: PMC7893827 DOI: 10.12659/msm.928455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/30/2020] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The IRF5 and TYK2 gene polymorphisms are associated with autoimmune diseases. However, the relationship between the IRF5 and TYK2 gene polymorphisms and RA risk in the Chinese Han population was inconsistent. MATERIAL AND METHODS A total of 578 RA patients (case group) and 578 healthy controls (control group) were assessed in a case-control study. Genotyping of IRF5 (Exon 6 insertion/deletion (in/de), rs2004640, rs2070197, rs10954213) and TYK2 (rs280500, rs280519, rs280521, rs8108236, rs12720253) was performed by direct sequencing method. Data analysis was performed by SHEsis. RESULTS The rs2004640T allele (P=0.0003) and the dominant (P=0.001) and recessive (P=0.01) models of rs2004640 were associated with RA risk after stringent Bonferroni correction (0.05/4). The IRF5 exon 6 (in), rs2070197 and rs10954213 were not associated with RA (P>0.05). Two haplotypes of IRF5 (DTAT and DTGG) were associated with RA susceptibility (P<0.05). In addition, the frequencies of TYK2 rs280500A, rs280521A, and rs8108236A were significantly higher in the RA group compared with the control group (P<0.05). TYK2 rs280500, rs280521, and rs8108236 were associated with RA susceptibility in the dominant model, but the same was not observed for rs280519 and rs12720253 (P<0.05). Furthermore, 3 risk haplotypes (AAAGT, AGGAT, and GAAAT) and a protective haplotype (GAGGT) of TYK2 gene were associated with RA susceptibility (P<0.05). CONCLUSIONS Our results suggest that IRF5 rs2004640, TYK2 rs280500, rs280521, rs8108236, and haplotypes IRF5 (DTAT and DTGG) and TYK2 (AAAGT, AGGAT, GAAAT, and GAGGT) are susceptible factors for RA in a Chinese Han population.
Collapse
|
28
|
A Comparison of Gene Expression Changes in the Blood of Individuals Consuming Diets Supplemented with Olives, Nuts or Long-Chain Omega-3 Fatty Acids. Nutrients 2020; 12:nu12123765. [PMID: 33302351 PMCID: PMC7762614 DOI: 10.3390/nu12123765] [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: 11/13/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
Background: The Mediterranean diet, which is rich in olive oil, nuts, and fish, is considered healthy and may reduce the risk of chronic diseases. Methods: Here, we compared the transcriptome from the blood of subjects with diets supplemented with olives, nuts, or long-chain omega-3 fatty acids and identified the genes differentially expressed. The dietary genes obtained were subjected to network analysis to determine the main pathways, as well as the transcription factors and microRNA interaction networks to elucidate their regulation. Finally, a gene-associated disease interaction network was performed. Results: We identified several genes whose expression is altered after the intake of components of the Mediterranean diets compared to controls. These genes were associated with infection and inflammation. Transcription factors and miRNAs were identified as potential regulators of the dietary genes. Interestingly, caspase 1 and sialophorin are differentially expressed in the opposite direction after the intake of supplements compared to Alzheimer’s disease patients. In addition, ten transcription factors were identified that regulated gene expression in supplemented diets, mild cognitive impairment, and Alzheimer’s disease. Conclusions: We identified genes whose expression is altered after the intake of the supplements as well as the transcription factors and miRNAs involved in their regulation. These genes are associated with schizophrenia, neoplasms, and rheumatic arthritis, suggesting that the Mediterranean diet may be beneficial in reducing these diseases. In addition, the results suggest that the Mediterranean diet may also be beneficial in reducing the risk of dementia.
Collapse
|
29
|
Ching YM, Viswanathan S, Mohamed Nor N, Shuib S, Kamarudin B, Mansor S, Yusof AY, Arip M. Association of CD58 polymorphism and multiple sclerosis in Malaysia: a pilot study. AUTOIMMUNITY HIGHLIGHTS 2020; 10:13. [PMID: 32257069 PMCID: PMC7065364 DOI: 10.1186/s13317-019-0123-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/06/2019] [Indexed: 11/15/2022]
Abstract
Background Multiple sclerosis is an immune mediated disease targeting the central nervous system. Association of non-human leukocyte antigen gene, CD58, with multiple sclerosis has been reported in several populations but is unclear among Southeast Asians. This pilot study was conducted to explore the association between CD58 polymorphism and multiple sclerosis among the Malay population in Malaysia. Methods Blood samples were collected from 27 multiple sclerosis patients, and compared with 58 age- and gender matched healthy individuals. All patients were tested negative for anti-aquaporin 4. DNA was extracted from the blood and genotyped for 3 single nucleotide polymorphisms rs12044852, rs2300747 and rs1335532 of gene CD58 by real-time PCR. Results The majority of multiple sclerosis patients were female (85.2%). The general mean age of onset was 30.5 years. Genotyping results showed that frequencies of the alleles were between 40 and 50% for MS patients and healthy individuals. Association (allelic model) between multiple sclerosis and CD58 gene polymorphism alleles rs12044852 (p = 0.410), rs2300747 (p = 0.881) and rs1335532 (p = 0.407) were indistinct. Conclusions The impact of the CD58 gene polymorphism was not prominent in this pilot study, implying that genetic composition contributing to multiple sclerosis may be different between different populations, thus results in a heterogeneity of disease manifestation and distribution.
Collapse
Affiliation(s)
- Yee Ming Ching
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | | | - Nurhanani Mohamed Nor
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Shuwahida Shuib
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Balqis Kamarudin
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Salawati Mansor
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Ainur Yusniza Yusof
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Masita Arip
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia.,3Allergy and Immunology Research Centre, Institute for Medical Research, Block C6, National Institute of Health Complex, No. 1, Jalan Setia Murni U13/52, Seksyen U13, Bandar Setia Alam, 40170 Shah Alam, Selangor Darul Ehsan Malaysia
| |
Collapse
|
30
|
Laufer VA, Tiwari HK, Reynolds RJ, Danila MI, Wang J, Edberg JC, Kimberly RP, Kottyan LC, Harley JB, Mikuls TR, Gregersen PK, Absher DM, Langefeld CD, Arnett DK, Bridges SL. Genetic influences on susceptibility to rheumatoid arthritis in African-Americans. Hum Mol Genet 2020; 28:858-874. [PMID: 30423114 DOI: 10.1093/hmg/ddy395] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/05/2018] [Accepted: 11/09/2018] [Indexed: 12/29/2022] Open
Abstract
Large meta-analyses of rheumatoid arthritis (RA) susceptibility in European (EUR) and East Asian (EAS) populations have identified >100 RA risk loci, but genome-wide studies of RA in African-Americans (AAs) are absent. To address this disparity, we performed an analysis of 916 AA RA patients and 1392 controls and aggregated our data with genotyping data from >100 000 EUR and Asian RA patients and controls. We identified two novel risk loci that appear to be specific to AAs: GPC5 and RBFOX1 (PAA < 5 × 10-9). Most RA risk loci are shared across different ethnicities, but among discordant loci, we observed strong enrichment of variants having large effect sizes. We found strong evidence of effect concordance for only 3 of the 21 largest effect index variants in EURs. We used the trans-ethnic fine-mapping algorithm PAINTOR3 to prioritize risk variants in >90 RA risk loci. Addition of AA data to those of EUR and EAS descent enabled identification of seven novel high-confidence candidate pathogenic variants (defined by posterior probability > 0.8). In summary, our trans-ethnic analyses are the first to include AAs, identified several new RA risk loci and point to candidate pathogenic variants that may underlie this common autoimmune disease. These findings may lead to better ways to diagnose or stratify treatment approaches in RA.
Collapse
Affiliation(s)
- Vincent A Laufer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hemant K Tiwari
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard J Reynolds
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Maria I Danila
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jelai Wang
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeffrey C Edberg
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert P Kimberly
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Leah C Kottyan
- Center for Autoimmune Genetics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - John B Harley
- Center for Autoimmune Genetics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,United States Department of Veterans Affairs Medical Center, Cincinnati, OH, USA
| | - Ted R Mikuls
- VA Nebraska-Western Iowa Health Care System and the Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter K Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY, USA
| | - Devin M Absher
- Hudson Alpha Institute for Biotechnology, Huntsville, AL, USA
| | - Carl D Langefeld
- Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Donna K Arnett
- University of Kentucky College of Public Health, Lexington, KY, USA
| | - S Louis Bridges
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
31
|
Abstract
Multiple sclerosis (MS) has a strong racial and ethnic component and disproportionately affects whites of European background. Recent incidence reports suggest an increasing rate of MS among African Americans compared with whites. Despite this recent increase in MS in African Americans, Hispanics and Asians are significantly less likely to develop MS than whites of European ancestry. MS-specific mortality trends demonstrate distinctive disparities by race/ethnicity and age, suggesting that there is an unequal burden of disease. Inequalities in health along with differences in clinical characteristics that may be genetic, environmental, and social in origin may be contributing to disease variability and be suggestive of endophenotypes. The overarching goal of this review was to summarize the current understanding on the variability of disease that we observe in selected racial and ethnic populations: Hispanics and African Americans. Future challenges will be to unravel the genetic, environmental, and social determinants of the observed racial/ethnic disparities.
Collapse
Affiliation(s)
- Lilyana Amezcua
- Multiple Sclerosis Comprehensive Care Center, University of Southern California, Los Angeles, CA, USA/Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jacob L McCauley
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
32
|
Dysregulation of a specific immune-related network of genes biologically defines a subset of schizophrenia. Transl Psychiatry 2019; 9:156. [PMID: 31150013 PMCID: PMC6544656 DOI: 10.1038/s41398-019-0486-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 03/22/2019] [Accepted: 04/29/2019] [Indexed: 12/31/2022] Open
Abstract
Currently, the clinical diagnosis of schizophrenia relies solely on self-reporting and clinical interview, and likely comprises heterogeneous biological subsets. Such subsets may be defined by an underlying biology leading to solid biomarkers. A transgenic rat model modestly overexpressing the full-length, non-mutant Disrupted-in-Schizophrenia 1 (DISC1) protein (tgDISC1 rat) was generated that defines such a subset, inspired by our previous identification of insoluble DISC1 protein in post mortem brains from patients with chronic mental illness. Besides specific phenotypes such as DISC1 protein pathology, abnormal dopamine homeostasis, and changes in neuroanatomy and behavior, this animal model also shows subtle disturbances in overarching signaling pathways relevant for schizophrenia. In a reverse-translational approach, assuming that both the animal model and a patient subset share common disturbed signaling pathways, we identified differentially expressed transcripts from peripheral blood mononuclear cells of tgDISC1 rats that revealed an interconnected set of dysregulated genes, led by decreased expression of regulator of G-protein signaling 1 (RGS1), chemokine (C-C) ligand 4 (CCL4), and other immune-related transcripts enriched in T-cell and macrophage signaling and converging in one module after weighted gene correlation network analysis. Testing expression of this gene network in two independent cohorts of patients with schizophrenia versus healthy controls (n = 16/50 and n = 54/45) demonstrated similar expression changes. The two top markers RGS1 and CCL4 defined a subset of 27% of patients with 97% specificity. Thus, analogous aberrant signaling pathways can be identified by a blood test in an animal model and a corresponding schizophrenia patient subset, suggesting that in this animal model tailored pharmacotherapies for this patient subset could be achieved.
Collapse
|
33
|
Tissier R, Hocini H, Tchitchek N, Deye N, Legriel S, Pichon N, Daubin C, Hermine O, Carli P, Vivien B, Tréluyer JM, Lefebvre C, Tisserand P, Dubois-Randé JL, Berdeaux A, Ghaleh B, Lelièvre JD, Levy Y, Cariou A. Early blood transcriptomic signature predicts patients' outcome after out-of-hospital cardiac arrest. Resuscitation 2019; 138:222-232. [PMID: 30885824 DOI: 10.1016/j.resuscitation.2019.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/27/2019] [Accepted: 03/10/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Early prognostication is a major challenge after out-of-hospital cardiac arrest (OHCA). AIMS We hypothesized that a genome-wide analysis of blood gene expression could offer new prognostic tools and lines of research. METHODS Sixty-nine patients were enrolled from an ancillary study of the clinical trial NCT00999583 that tested the effect of erythropoietin (EPO) after OHCA. Blood samples were collected in comatose survivors of OHCA at hospital admission and 1 and 3 days after resuscitation. Gene expression profiles were analyzed (Illumina HumanHT-12 V4 BeadChip; >34,000 genes). Patients were classified into two categories representing neurological favorable outcome (cerebral performance category [CPC] = 1-2) vs unfavorable outcome (CPC > 2) at Day 60 after OHCA. Differential and functional enrichment analyses were performed to compare transcriptomic profiles between these two categories. RESULTS Among the 69 enrolled patients, 33 and 36 patients were treated or not by EPO, respectively. Among them, 42% had a favorable neurological outcome in both groups. EPO did not affect the transcriptomic response at Day-0 and 1 after OHCA. In contrast, 76 transcripts differed at Day-0 between patients with unfavorable vs favorable neurological outcome. This signature persisted at Day-1 after OHCA. Functional enrichment analysis revealed a down-regulation of adaptive immunity with concomitant up-regulation of innate immunity and inflammation in patients with unfavorable vs favorable neurological outcome. The transcription of many genes of the HLA family was decreased in patients with unfavorable vs favorable neurological outcome. Concomitantly, neutrophil activation and inflammation were observed. Up-stream regulators analysis showed the implication of numerous factors involved in cell cycle and damages. A logistic regression including a set of genes allowed a reliable prediction of the clinical outcomes (specificity = 88%; Hit Rate = 83%). CONCLUSIONS A transcriptomic signature involving a counterbalance between adaptive and innate immune responses is able to predict neurological outcome very early after hospital admission after OHCA. This deserves confirmation in a larger population.
Collapse
Affiliation(s)
- Renaud Tissier
- Inserm, U955, F94000, Créteil, France; Université Paris Est, UMR_S955, UPEC, Ecole Nationale Vétérinaire d'Alfort, F-94000, Créteil, France.
| | - Hakim Hocini
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France
| | - Nicolas Tchitchek
- Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France; CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure, F-92265 Fontenay-aux-Roses, France
| | - Nicolas Deye
- Medical ICU, Inserm U942, Lariboisiere Hospital, APHP, F-75010, Paris, France
| | - Stéphane Legriel
- Intensive Care Unit, Versailles Hospital, Le Chesnay, F-78150, France
| | - Nicolas Pichon
- Intensive Care Unit, University Hospital Dupuytren, Limoges, F-87042, France
| | - Cédric Daubin
- CHU de Caen, Department of Medical Intensive Care, Caen, F-14000, France
| | - Olivier Hermine
- Department of Hematology and INSERM U1163 CNRS ERL 8654, Imagine Institute and Necker Hospital, Paris, F-75015, France
| | - Pierre Carli
- SAMU de Paris, Service d'Anesthésie-Réanimation, Hôpital Universitaire Necker- Enfants Malades, Université Paris Descartes, F-75015, Paris, France
| | - Benoît Vivien
- SAMU de Paris, Service d'Anesthésie-Réanimation, Hôpital Universitaire Necker- Enfants Malades, Université Paris Descartes, F-75015, Paris, France
| | - Jean-Marc Tréluyer
- Clinical Research Unit, Paris Centre and Paris Descartes University, Paris, France
| | - Cécile Lefebvre
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France
| | - Pascaline Tisserand
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France
| | - Jean-Luc Dubois-Randé
- Inserm, U955, F94000, Créteil, France; Université Paris Est, UMR_S955, UPEC, Ecole Nationale Vétérinaire d'Alfort, F-94000, Créteil, France
| | - Alain Berdeaux
- Inserm, U955, F94000, Créteil, France; Université Paris Est, UMR_S955, UPEC, Ecole Nationale Vétérinaire d'Alfort, F-94000, Créteil, France
| | - Bijan Ghaleh
- Inserm, U955, F94000, Créteil, France; Université Paris Est, UMR_S955, UPEC, Ecole Nationale Vétérinaire d'Alfort, F-94000, Créteil, France; APHP, Hôpitaux Universitaires Henri Mondor, Plateforme de Ressources Biologiques, F-94000, Créteil, France
| | - Jean-Daniel Lelièvre
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France
| | - Yves Levy
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France.
| | - Alain Cariou
- Service de Réanimation Médicale, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Paris, France
| |
Collapse
|
34
|
Kulakova OG, Bashinskaya VV, Tsareva EY, Boyko AN, Favorova OO, Gusev EI. [Association analysis of cytokine receptors' genes polymorphisms with clinical features of multiple sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 116:10-15. [PMID: 28139605 DOI: 10.17116/jnevro201611610210-15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AIM To study the association of polymorphisms in the IL2RA and TNFRSF1A genes with severity and early clinical manifestations of remitted multiple sclerosis (MS). MATERIAL AND METHODS Five hundred and eight patients of Russian ethnicity with bout-onset MS were genotyped for IL7RA (rs6897932), IL2RA (rs2104286) and TNFRSF1A (rs1800693) polymorphisms. Association analysis of the gene variants with disease severity, variants of MS manifestation, and first remission duration was performed. RESULTS AND CONCLUSION Dividing the MS patients by disease severity, estimated with the MSSS, we found a significant increase in the TNFRSF1A*T/T genotype carriage in patients with milder MS course (MSSS≤3), and, respectively, in the TNFRSF1A*C allele carriage in patients with moderate to severe MS (MSSS> 3). Dividing the MS patients into two groups according to their MS manifestation variants, we revealed a significant increase in the TNFRSF1A*T allele carriage in patients with favorable variants of MS manifestation (optic neuritis or sensory disturbances), and of the TNFRSF1A*C/C genotype in patients with unfavorable variants (motor disorders, brain stem disorders, impaired coordination, pelvic disorders, mental disorders or polysymptomatic onset). No associations with first remission duration were observed. Multi-locus analysis to search for allelic combinations associated with the studied clinical features of MS was applied. In this analysis, a polymorphic variant of CTLA4 gene (rs231775), for which we have previously reported the association of the CTLA4*G allele with short first remission (less than 1 year), was also included. The carriage of biallelic combination (CTLA4*G + TNFRSF1A*C) was associated with short first remission more significantly than the carriage of CTLA4*G by itself. One more biallelic combination associated with short first remission (CTLA4*G/G + IL7RA*T), was identified. No other biallelic combinations significantly associated with the clinical features studied were observed.
Collapse
Affiliation(s)
- O G Kulakova
- Pirogov Russian National Research Medical University, Moscow, Russia; Russian Cardiology Research and Production Center, Moscow, Russia
| | - V V Bashinskaya
- Pirogov Russian National Research Medical University, Moscow, Russia; Russian Cardiology Research and Production Center, Moscow, Russia
| | - E Yu Tsareva
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A N Boyko
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - O O Favorova
- Pirogov Russian National Research Medical University, Moscow, Russia; Russian Cardiology Research and Production Center, Moscow, Russia
| | - E I Gusev
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
35
|
Petracca M, Zaaraoui W, Cocozza S, Vancea R, Howard J, Heinig MM, Fleysher L, Oesingmann N, Ranjeva JP, Inglese M. An MRI evaluation of grey matter damage in African Americans with MS. Mult Scler Relat Disord 2018; 25:29-36. [PMID: 30029018 PMCID: PMC6214725 DOI: 10.1016/j.msard.2018.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/29/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Multiple sclerosis (MS) is less prevalent in African Americans (AAs) than Caucasians (CAs) but in the former the disease course tends to be more severe. In order to clarify the MRI correlates of disease severity in AAs, we performed a multimodal brain MRI study to comprehensively assess the extent of grey matter (GM) damage and the degree of functional adaptation to structural damage in AAs with MS. METHODS In this cross-sectional study, we characterized GM damage in terms of focal lesions and volume loss and functional adaptation during the execution of a simple motor task on a sample of 20 AAs and 20 CAs with MS and 20 healthy controls (CTRLs). RESULTS In AAs, we observed a wider range of EDSS scores than CAs, with multisystem involvement being more likely in AAs (p < 0.01). While no significant differences were detected in lesion loads and global brain volumes, AAs showed regional atrophy in the posterior lobules of cerebellum, temporo-occipital and frontal regions in comparison with CAs (p < 0.01), with cerebellar atrophy being the best metric in differentiating AAs from CAs (p = 0.007, AUC = 0.96 and p = 0.005, AUC = 0.96, respectively for right and left cerebellar clusters). In AAs, the functional analysis of cortical activations showed an increase in task-related activation of areas involved in high level processing and a decreased activation in the medial prefrontal cortex compared to CAs. INTERPRETATION In our study, the direct comparison of AAs and CAs points to cerebellar atrophy as the main difference between subgroups.
Collapse
Affiliation(s)
- Maria Petracca
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Neurosciences, Reproductive and Odonto-stomatological Sciences, University "Federico II", Naples, Italy
| | - Wafaa Zaaraoui
- Aix-Marseille Univ, CNRS, CRMBM UMR, 7339, Marseille, France
| | - Sirio Cocozza
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Roxana Vancea
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jonathan Howard
- Department of Neurology, New York University School of Medicine, New York, NY, USA
| | - Monika M Heinig
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Lazar Fleysher
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | | | - Jean-Philippe Ranjeva
- Aix-Marseille Univ, CNRS, CRMBM UMR, 7339, Marseille, France; UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| | - Matilde Inglese
- Department of Neurology, Radiology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, (DINOGMI) University of Genova and IRCCS AOU San Martino-IST, Genoa, Italy.
| |
Collapse
|
36
|
Zaplakhova OV, Nasibullin TR, Tuktarova IA, Timasheva YR, Erdman VV, Bakhtiyarova KZ, Mustafina OE. Associations of Polymorphic DNA Markers and Their Combinations with Multiple Sclerosis. RUSS J GENET+ 2018. [DOI: 10.1134/s102279541808015x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Javor J, Shawkatová I, Ďurmanová V, Párnická Z, Čierny D, Michalik J, Čopíková-Cudráková D, Smahová B, Gmitterová K, Peterajová Ľ, Bucová M. TNFRSF1A polymorphisms and their role in multiple sclerosis susceptibility and severity in the Slovak population. Int J Immunogenet 2018; 45:257-265. [PMID: 30009568 DOI: 10.1111/iji.12388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/24/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022]
Abstract
Tumour necrosis factor (TNF)-mediated signalling plays a key role in inflammatory and neurodegenerative processes leading to the development of multiple sclerosis (MS). Recent studies have highlighted the role of tumour necrosis factor receptor superfamily member 1A (TNFRSF1A) gene encoding the type 1 TNF receptor in the genetic predisposition to MS. This study aimed to validate the association of TNFRSF1A rs1800693 and rs4149584 polymorphisms with susceptibility to MS in the Slovak population and analyse their influence on age at disease onset, severity, and disability progression. Polymerase chain reaction-restriction fragment length polymorphism method was used to genotype both TNFRSF1A polymorphisms in 541 MS patients and 724 healthy controls. Logistic regression analysis revealed a significantly increased risk of developing MS for the carriers of rs1800693 C allele (TC + CC vs. TT: pcorr = 0.005; OR = 1.61; 95% CI = 1.23-2.12), irrespective of sex and carriage of the major MS risk allele HLA-DRB1*15:01. On the other hand, no association could be found between rs4149584 and MS risk (GA + AA vs. GG: pcorr = 1.00; OR = 1.25; 95% CI = 0.71-2.21). Moreover, neither polymorphism was significantly associated with age at disease onset, MS Severity Score (MSSS) or MS Progression Index (PI) in any of the inheritance models. In conclusion, our results provide support for a sex- and HLA-DRB1*15:01-independent association of TNFRSF1A rs1800693 SNP with MS susceptibility, but not with age at disease onset, severity or rate of disability accumulation.
Collapse
Affiliation(s)
- Juraj Javor
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Ivana Shawkatová
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Vladimíra Ďurmanová
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Zuzana Párnická
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Daniel Čierny
- Department of Clinical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava and University Hospital Martin, Martin, Slovakia
| | - Jozef Michalik
- Clinic of Neurology, Jessenius Faculty of Medicine, Comenius University in Bratislava and University Hospital Martin, Martin, Slovakia
| | - Daniela Čopíková-Cudráková
- 1st Department of Neurology, Faculty of Medicine, Comenius University in Bratislava and University Hospital Bratislava, Bratislava, Slovakia
| | - Barbora Smahová
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Karin Gmitterová
- 2nd Department of Neurology, Faculty of Medicine, Comenius University in Bratislava and University Hospital Bratislava, Bratislava, Slovakia
| | - Ľubica Peterajová
- Haematology Outpatient Clinic, University Hospital Bratislava, Bratislava, Slovakia
| | - Mária Bucová
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
38
|
Abstract
The contribution of genetic inheritance in multiple sclerosis was established early on. Although multiple sclerosis is not a Mendelian disease, its incidence and prevalence is higher in family members of affected individuals compared with the general population. Throughout the last decade, several small studies failed to identify any robust genetic associations besides the classic associations in the major histocompatibility complex region. During the past few years, genome-wide association studies (GWAS) have revolutionized the genetics of multiple sclerosis, uncovering more than 200 implicated genetic loci. Here, we describe these main findings and discuss the new avenues that these discoveries lay open.
Collapse
Affiliation(s)
- Nikolaos A Patsopoulos
- Department of Neurology, Brigham & Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts 02142
| |
Collapse
|
39
|
Squires KE, Montañez-Miranda C, Pandya RR, Torres MP, Hepler JR. Genetic Analysis of Rare Human Variants of Regulators of G Protein Signaling Proteins and Their Role in Human Physiology and Disease. Pharmacol Rev 2018; 70:446-474. [PMID: 29871944 PMCID: PMC5989036 DOI: 10.1124/pr.117.015354] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulators of G protein signaling (RGS) proteins modulate the physiologic actions of many neurotransmitters, hormones, and other signaling molecules. Human RGS proteins comprise a family of 20 canonical proteins that bind directly to G protein-coupled receptors/G protein complexes to limit the lifetime of their signaling events, which regulate all aspects of cell and organ physiology. Genetic variations account for diverse human traits and individual predispositions to disease. RGS proteins contribute to many complex polygenic human traits and pathologies such as hypertension, atherosclerosis, schizophrenia, depression, addiction, cancers, and many others. Recent analysis indicates that most human diseases are due to extremely rare genetic variants. In this study, we summarize physiologic roles for RGS proteins and links to human diseases/traits and report rare variants found within each human RGS protein exome sequence derived from global population studies. Each RGS sequence is analyzed using recently described bioinformatics and proteomic tools for measures of missense tolerance ratio paired with combined annotation-dependent depletion scores, and protein post-translational modification (PTM) alignment cluster analysis. We highlight selected variants within the well-studied RGS domain that likely disrupt RGS protein functions and provide comprehensive variant and PTM data for each RGS protein for future study. We propose that rare variants in functionally sensitive regions of RGS proteins confer profound change-of-function phenotypes that may contribute, in newly appreciated ways, to complex human diseases and/or traits. This information provides investigators with a valuable database to explore variation in RGS protein function, and for targeting RGS proteins as future therapeutic targets.
Collapse
Affiliation(s)
- Katherine E Squires
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Carolina Montañez-Miranda
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Rushika R Pandya
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Matthew P Torres
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - John R Hepler
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| |
Collapse
|
40
|
Tizaoui K. Multiple sclerosis genetics: Results from meta-analyses of candidate-gene association studies. Cytokine 2018; 106:154-164. [DOI: 10.1016/j.cyto.2017.10.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/16/2017] [Accepted: 10/25/2017] [Indexed: 12/12/2022]
|
41
|
Rivas-Rodríguez E, Amezcua L. Ethnic Considerations and Multiple Sclerosis Disease Variability in the United States. Neurol Clin 2018; 36:151-162. [DOI: 10.1016/j.ncl.2017.08.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
42
|
Panezai J, Ghaffar A, Altamash M, Sundqvist KG, Engström PE, Larsson A. Correlation of serum cytokines, chemokines, growth factors and enzymes with periodontal disease parameters. PLoS One 2017; 12:e0188945. [PMID: 29190740 PMCID: PMC5708747 DOI: 10.1371/journal.pone.0188945] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/15/2017] [Indexed: 01/11/2023] Open
Abstract
Background Periodontal disease (PD) is characterized by inflammatory tissue destruction in tooth supporting apparatus. Many studies indicate that the underlying pathogenesis is in concordance with rheumatoid arthritis (RA) sharing immune-inflammatory events affect both diseases. The aim of this study was to investigate serum cytokines, chemokines, growth factors, enzymes and costimulatory proteins in association with periodontal conditions in PD and RA subjects. Materials & methods Periodontal examination was performed in RA (n = 38), PD (n = 38) and healthy subjects (n = 14). Bleeding on probing (BOP) and probing pocket depth (PPD) were measured. Marginal bone loss (MBL) for premolars and molars was measured on digital panoramic radiographs. PD was defined as present if the PPD was ≥5mm in ≥ 3 different sites. Serum samples were collected from all subjects. A multiplex proximity extension assay (PEA) was used to analyze the samples for simultaneous measurement of 92 cytokines. Cytokines with ≥ 60% quantitative results were included. Results A significant positive correlation was seen for ST1A1, FGF-19 and NT-3 whereas EN-RAGE, DNER, CX3CL1 and TWEAK associated inversely with BOP, PPD≥ 5mm and MBL but positively with number of teeth. Several CD markers (CD244, CD40, CDCP1, LIF-R, IL-10RA, CD5 and CD6) were found to be associated with BOP, shallow and deep pockets, MBL and number of teeth, either directly or inversely. Most chemokines (CCL8, CX3CL1, CXCL10, CXCL11, CCL11, CCL4, CCL20, CXCL5, CXCL6, and CCL23) were positively associated with number of teeth and some inversely related to MBL (CCL8, CXCL10). Proteins with enzymatic activity (ST1A1, HGF and CASP-8) were directly related to the severity of periodontal conditions and inversely related to number of teeth. Aside from FGF-19, other growth factors were also directly associated with MBL (HGF), number of teeth (VEGF-A, LAP TGF-beta-1) and, inversely to, shallow pockets (LAP TGF-beta-1, TGFA and Beta-NGF). Out of 33 cytokines, 32 associated inversely with shallow pockets, whereas only CD40 associated positively. Associations between cytokines and periodontal parameters in the RA group were comparatively less. Statistical analyses were adjusted for multivariate effects using the Benjamini–Hochberg false discovery rate method. Conclusion Systemic inflammatory burden, via known and novel markers, is associated with periodontal conditions in PD and RA subjects. Shallow pockets are not associated with a higher inflammatory state.
Collapse
Affiliation(s)
- Jeneen Panezai
- Altamash Institute of Dental Medicine, Department of Periodontology, Karachi, Pakistan
- Karolinska Institutet, Department of Dental Medicine, Division of Periodontology, Huddinge, Sweden
- * E-mail:
| | - Ambereen Ghaffar
- Habib Medical Centre, Rheumatology Clinic, Karachi, Pakistan, Karachi, Pakistan
| | - Mohammad Altamash
- Altamash Institute of Dental Medicine, Department of Periodontology, Karachi, Pakistan
| | - Karl-Gösta Sundqvist
- Karolinska University Hospital, Department of Laboratory Medicine, Division of Clinical Immunology, Stockholm, Sweden
| | - Per-Erik Engström
- Karolinska Institutet, Department of Dental Medicine, Division of Periodontology, Huddinge, Sweden
| | - Anders Larsson
- Uppsala University, Department of Medical Sciences, Uppsala, Sweden
| |
Collapse
|
43
|
Genetic Factors Associated with Risk and Disability Progression of Multiple Sclerosis in Slovak Population. ACTA MEDICA MARTINIANA 2017. [DOI: 10.1515/acm-2017-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Objective: The aim of our study was to determine the relation of particular genetic variants in selected genes (GSTM1, GSTT1 null genotypes; rs1695 GSTP1; rs10735781 EVI5) to the risk of multiple sclerosis (MS) development and find out the possible association with disease disability progression rate. Material and methods: Our study included 202 MS patients and 174 healthy control volunteers. MS patients were divided according to disability progression rate to three groups - slowly progressing, mid-rate progressing and rapidly progressing. All DNA samples were isolated from venous blood. Genotyping was performed by PCR-RFLP and multiplex PCR. Results: Our analysis showed that GSTT1 null genotype (OR 0.56; 95%CI 0.33 -0.95; p=0.04) and GSTM1, GSTT1 double null genotype (OR 0.32; 95%CI 0.14 - 0.74; p=0.006) are potentially protective in relation to MS. We observed similar result in GSTT1 null genotype in association with mid-rate progression (OR 0.48; 95%CI 0.24 - 0.97; p=0.05). Frequency of GSTM1 and GSTT1 double null genotype is significantly lower in subgroup of MS patients with progression rate defined as slow (OR 0.22; 95%CI 0.05 - 0.98; p=0.05) and middle (OR 0.33; 95%CI 0.11 - 0.99; p=0.045). We did not show any significant association of genetic changes rs1695 in GSTP1 and rs10735781 in EVI5 with MS or rate of disease progression. Conclusions: Genetic basis of multiple sclerosis is still not fully elucidated. Further research may clarify our results and confirm the value of studied factors for clinical practice.
Collapse
|
44
|
Ecotropic Viral Integration Site 5 (EVI5) variants are associated with multiple sclerosis in Iranian population. Mult Scler Relat Disord 2017; 18:15-19. [PMID: 29141798 DOI: 10.1016/j.msard.2017.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/21/2017] [Accepted: 09/11/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a multifactorial disorder with immunological basis. Numerous genetic and environmental factors contribute in its pathogenesis. Several genetic loci have been shown to be associated with MS risk. Among genes whose participation in MS has been evaluated is Ecotropic Viral Integration Site 5 (EVI5). EVI5 is a common site of retroviral integration with a possible role in T-cell lymphomagenesis. METHODS In the current study, we aimed to confirm association of the single nucleotide polymorphisms (SNPs) within EVI5 gene with MS in 410 relapsing-remitting MS patients and 410 controls from Iranian population. The rs6680578, rs10537781 and rs11810217 genotypes were defined by amplification-refractory mutation system (ARMS)-PCR method. RESULTS The allele and genotype frequencies of rs6680578 and rs11810217 were not significantly different between cases and controls. However, in the rs10735781 the GG genotype was significantly associated with MS risk in recessive (P = 0.03, OR (95%CI) = 1.84 (1.05-3.19)) and co-dominant models (P = 0.02, OR 95%CI) = 1.90 (1.08-3.35)). In addition, T G T haplotype (rs6680578, rs10735781 and rs11810217 respectively) was associated with MS risk while T C C, A G T and A C T had a protective effect against MS. CONCLUSION The results of the current study provide further evidences for participation of EVI5 in MS pathogenesis.
Collapse
|
45
|
Zaplakhova OV, Timasheva YR, Bakhtiyarova KZ, Tuktarova IA, Mustafina OE. [Clinical and molecular genetic analysis of a case of familial multiple sclerosis in the Republic of Bashkortostan]. Zh Nevrol Psikhiatr Im S S Korsakova 2017; 117:31-41. [PMID: 28617359 DOI: 10.17116/jnevro20171172231-41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
AIM To investigate clinical manifestations of multiple sclerosis (MS) and the genetic makeup of six affected members of one family. MATERIAL AND METHODS Six members of the family of Russian ethnic origin were examined. Pedigree analysis and genotyping of polymorphic markers of candidate genes for multiple sclerosis were performed. RESULTS AND CONCLUSION The accumulation of alleles that were associated with autoimmune diseases according to the results of genome-wide association studies (rs1109670*C, rs3129934*T, rs9523762*G, rs1570538*T) was found in the family. The results confirm the contribution of several genetic variants to familial forms of MS.
Collapse
Affiliation(s)
- O V Zaplakhova
- Institute of Biochemistry and Genetics Ufa Scientific Centre of Russian Academy of Sciences, Ufa, Russia; Bashkir State Medical University, Ufa, Russia
| | - Ya R Timasheva
- Institute of Biochemistry and Genetics Ufa Scientific Centre of Russian Academy of Sciences, Ufa, Russia
| | | | - I A Tuktarova
- Institute of Biochemistry and Genetics Ufa Scientific Centre of Russian Academy of Sciences, Ufa, Russia
| | - O E Mustafina
- Institute of Biochemistry and Genetics Ufa Scientific Centre of Russian Academy of Sciences, Ufa, Russia
| |
Collapse
|
46
|
Majoros A, Platanitis E, Kernbauer-Hölzl E, Rosebrock F, Müller M, Decker T. Canonical and Non-Canonical Aspects of JAK-STAT Signaling: Lessons from Interferons for Cytokine Responses. Front Immunol 2017; 8:29. [PMID: 28184222 PMCID: PMC5266721 DOI: 10.3389/fimmu.2017.00029] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/09/2017] [Indexed: 01/07/2023] Open
Abstract
Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signal transduction mediates cytokine responses. Canonical signaling is based on STAT tyrosine phosphorylation by activated JAKs. Downstream of interferon (IFN) receptors, activated JAKs cause the formation of the transcription factors IFN-stimulated gene factor 3 (ISGF3), a heterotrimer of STAT1, STAT2 and interferon regulatory factor 9 (IRF9) subunits, and gamma interferon-activated factor (GAF), a STAT1 homodimer. In recent years, several deviations from this paradigm were reported. These include kinase-independent JAK functions as well as extra- and intranuclear activities of U-STATs without phosphotyrosines. Additionally, transcriptional control by STAT complexes resembling neither GAF nor ISGF3 contributes to transcriptome changes in IFN-treated cells. Our review summarizes the contribution of non-canonical JAK-STAT signaling to the innate antimicrobial immunity imparted by IFN. Moreover, we touch upon functions of IFN pathway proteins beyond the IFN response. These include metabolic functions of IRF9 as well as the regulation of natural killer cell activity by kinase-dead TYK2 and different phosphorylation isoforms of STAT1.
Collapse
Affiliation(s)
- Andrea Majoros
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Ekaterini Platanitis
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Elisabeth Kernbauer-Hölzl
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Felix Rosebrock
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Decker
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Aurenção JCK, Vasconcelos CCF, Thuler LCS, Alvarenga RMP. Disability and progression in Afro-descendant patients with multiple sclerosis. ARQUIVOS DE NEURO-PSIQUIATRIA 2016; 74:836-841. [DOI: 10.1590/0004-282x20160118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 05/25/2016] [Indexed: 11/21/2022]
Abstract
ABSTRACT Multiple sclerosis (MS) prevalence is higher in Caucasian (CA) populations, narrowing the analysis of the impact of Afro-descendant (AD) populations in disease outcomes. Even so, recent studies observed that AD patients have a more severe course. The main objective of this study is to confirm and discuss, through a systematic review, that being AD is a risk factor for disability accumulation and/or severe progression in patients with MS. A systematic review of published data in the last eleven years was performed, which evaluated clinical aspects and long term disability in patients with MS. Fourteen studies were included. Of these fourteen articles, thirteen observed a relationship between ancestry and poorer outcome of MS. African ancestry is a condition inherent in the patient and should be considered as an initial clinical characteristic affecting prognosis, and influencing which therapeutic decision to make in initial phases.
Collapse
|
48
|
Liu J, Liu X, Liu Y, Deng S, Huang H, Chen Q, Liu W, Huang Z. Association of EVI5 rs11808092, CD58 rs2300747, and CIITA rs3087456 polymorphisms with multiple sclerosis risk: A meta-analysis. Meta Gene 2016; 9:97-103. [PMID: 27331013 PMCID: PMC4908284 DOI: 10.1016/j.mgene.2016.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/19/2016] [Accepted: 04/22/2016] [Indexed: 11/15/2022] Open
Abstract
Purpose Multiple sclerosis (MS) is a major demyelinating disease of the central nervous system with a strong genetic component. Previous studies have reported that the association of EVI5 rs11808092, CD58 rs2300747, and CIITA rs3087456 polymorphisms with the susceptibility to MS. However, the results were inconsistent. Thus, we conducted this meta-analysis to provide a more accurate estimation of the association between any of these polymorphisms and MS risk. Methods The PubMed, Embase, Chinese National Knowledge Infrastructure, Wan Fang databases and MSGene were used to search all potentially relevant studies. The odds ratio (OR) with 95% confidence interval (CI) was used to investigate the associations between these three polymorphisms and MS risk. Results 16 independent case–control studies from 12 publications were finally included into this meta-analysis. The results showed that EVI5 rs11808092 polymorphism was related with increasing the development of MS under five genetic models (allelic: OR = 1.17, 95% CI = 1.10–1.24, P < 0.01; homozygous: OR = 1.37, 95% CI = 1.18–1.59, P < 0.01; heterozygous: OR = 1.16, 95% CI = 1.07–1.26, P < 0.01; recessive: OR = 1.28, 95% CI = 1.11–1.48, P < 0.01; and dominant: OR = 1.19, 95% CI = 1.11–1.48, P < 0.01). CD58 rs2300747 polymorphism was found to be associated with decreasing MS risk in three genetic models (allelic: OR = 0.86, 95% CI = 0.78–0.94, P < 0.01; heterozygous: OR = 0.85, 95% CI = 0.76–0.94, P < 0.01, and dominant: OR = 0.84, 95% CI = 0.76–0.93, P < 0.01). However, this meta-analysis indicated that CIITA rs3087456 polymorphism was not related to multiple sclerosis. Conclusions The mutant alleles of EVI5 rs11808092 polymorphism may increase the susceptibility to MS while those of CD58 rs2300747 polymorphism may decrease MS risk. In addition, CIITA rs3087456 polymorphism might not be associated with MS.
Collapse
Affiliation(s)
- Jiahe Liu
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China; The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Xu Liu
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China; The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Yong Liu
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China; School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China; Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, Zhanjiang, Guangdong 524023, China
| | - Shimin Deng
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China; School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Hongbin Huang
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China; The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Qicong Chen
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Weidong Liu
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China; School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Zunnan Huang
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China; School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China; Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, Zhanjiang, Guangdong 524023, China
| |
Collapse
|
49
|
Lee JK, Bou Dagher J. Regulator of G-protein Signaling (RGS)1 and RGS10 Proteins as Potential Drug Targets for Neuroinflammatory and Neurodegenerative Diseases. AAPS JOURNAL 2016; 18:545-9. [PMID: 26902301 DOI: 10.1208/s12248-016-9883-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/02/2016] [Indexed: 01/25/2023]
Abstract
Regulator of G-protein signaling (RGS) proteins were originally identified as negative regulators of G-protein-coupled receptor (GPCR) signaling via their GTPase-accelerating protein (GAP) activity. All RGS proteins contain evolutionarily conserved RGS domain; however, they differ in their size and regulatory domains. RGS1 and RGS10 are smaller than other RGS proteins, and their functions involve various inflammatory responses including autoimmune responses in both the periphery and the central nervous system (CNS). Neuroinflammation is the chronic inflammatory response in the CNS. Acute inflammatory response in the CNS is believed to be beneficial by involving the neuroprotective actions of immune cells in the brain, particularly microglia, to limit tissue damage and to aid in neuronal repair. However, chronically elevated levels of cytokines serve to maintain activation of abundant numbers of immune cells potentiating prolonged inflammatory responses and creating an environment of oxidative stress, which further hastens oxidative damage of neurons. In this review, we describe the implications and features of RGS proteins (specifically RGS1 and RGS10) in neuroinflammation and neurodegenerative diseases. We will discuss the experimental and epidemiological evidence on the benefits of anti-inflammatory interventions by targeting RGS1 and/or RGS10 protein function or expression in order to delay or attenuate the progression of neurodegeneration, particularly in multiple sclerosis (MS) and Parkinson's disease (PD).
Collapse
Affiliation(s)
- Jae-Kyung Lee
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Dr., Athens, Georgia, 30602, USA.
| | - Josephine Bou Dagher
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Dr., Athens, Georgia, 30602, USA
| |
Collapse
|
50
|
Wing AC, Hygino J, Ferreira TB, Kasahara TM, Barros PO, Sacramento PM, Andrade RM, Camargo S, Rueda F, Alves‐Leon SV, Vasconcelos CC, Alvarenga R, Bento CAM. Interleukin-17- and interleukin-22-secreting myelin-specific CD4(+) T cells resistant to corticoids are related with active brain lesions in multiple sclerosis patients. Immunology 2016; 147:212-20. [PMID: 26781085 PMCID: PMC4717237 DOI: 10.1111/imm.12552] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/16/2015] [Accepted: 10/27/2015] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is thought to be an autoimmune disorder. It is believed that immunological events in the early stages have great impact on the disease course. Therefore, we aimed to evaluate the cytokine profile of myelin basic protein (MBP)-specific T cells from MS patients in the early phase of the disease and correlate it to clinical parameters, as well as to the effect of in vitro corticoid treatment. Peripheral T cells from MS patients were stimulated with MBP with our without hydrocortisone for 5 days. The cytokines level were determined by ELISA. The number of active brain lesions was determined by MRI scans, and the neurological disabilities were assessed by Expanded Disability Status Scale scores. Our results demonstrated that MS-derived T cells responded to MBP by producing high levels of T helper type 1 (Th1) and Th17 cytokines. Although the production of interleukin-6 (IL-6), granulocyte-macrophage colony-stimulating factor, IL-17 and IL-22 was less sensitive to hydrocortisone inhibition, only IL-17 and IL-22 levels correlated with active brain lesions. The ability of hydrocortisone to inhibit IL-17 and IL-22 production by MBP-specific CD4(+) T cells was inversely related to the number of active brain lesions. Finally, the production of both cytokines was significantly higher in cell cultures from Afrodescendant patients and it was less sensitive to hydrocortisone inhibition. In summary, our data suggest that IL-17- and IL-22-secreting CD4(+) T cells resistant to corticoids are associated with radiological activity of the MS in early stages of the disease, mainly among Afrodescendant patients who, normally, have worse prognosis.
Collapse
Affiliation(s)
- Ana Cristina Wing
- Post‐graduate Programme in NeurologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Joana Hygino
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Thais B. Ferreira
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Taissa M. Kasahara
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Priscila O. Barros
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Priscila M. Sacramento
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Regis M. Andrade
- Department of General MedicineFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | | | - Fernanda Rueda
- Clinical Diagnosis by Image/Barra da Tijuca UnityRio de JaneiroBrazil
| | - Soniza V. Alves‐Leon
- Post‐graduate Programme in NeurologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | | | - Regina Alvarenga
- Post‐graduate Programme in NeurologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Cleonice A. M. Bento
- Post‐graduate Programme in NeurologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| |
Collapse
|