1
|
Rees K, Aicheler R, Butcher L, Dodd A, Geen J, Lynch C, Massey I, Morris K, Tennant B, Webb R. Seasonal variation in the associations between self-reported long-COVID symptoms and IL-6 signalling-related factors (particularly the rs2228145 variant of the IL-6R gene): A clinical study. Cytokine 2025; 189:156884. [PMID: 39987891 DOI: 10.1016/j.cyto.2025.156884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/25/2025]
Abstract
This observational study focused on the impact of Interleukin-6 (IL-6)-related factors (notably the IL-6 receptor (IL-6R) gene's rs2228145 polymorphism) on long-COVID risk in individuals who had previously experienced COVID-19 infection(s). The purpose of the study was to better understand such factors' contribution to long-COVID risk, and thus possibly initiate future strategies for using IL-6-related factors as biomarkers predictive of risk (while also obtaining data that may influence long-COVID management and treatment more generally). DNA and blood samples, plus questionnaire responses regarding long-COVID symptoms (including chronic fatigue and cognitive impairment), were collected from 175 participants who had previously experienced COVID-19 infection(s). Potential associations between self-reported long-COVID symptoms and participants' rs2228145 genotypes (determined using TaqMan-based genotyping assays) and/or their circulating IL-6, sIL-6R and sgp130 levels (determined using ELISA) were evaluated. Univariate-regression analyses demonstrated that odds of exhibiting long-COVID symptoms increased with severity/number of previous COVID-19 infection(s) and with hypertension as a co-morbidity, while vaccination decreased the likelihood of developing long-COVID. While long-COVID sufferers exhibited higher IL-6 signalling activity than healthy control individuals, rs2228145 genotype was not associated with long-COVID odds-ratios in- the entire-study cohort. Following identification of significant seasonal variations within our dataset, the entire-study cohort was stratified depending on when samples/questionnaire responses were obtained. In the resulting 'summer' sub-cohort (but not the 'winter' sub-cohort), the rs2228145 AA genotype was significantly over-represented amongst those exhibiting long-COVID symptoms, and long-COVID odds-ratios were significantly reduced for the CC and AC genotypes. While interpretation is complicated by seasonal variations, these findings may be of medical/biomedical value. Importantly, as IL-6 was higher in long-COVID sufferers than healthy controls, and rs2228145 AA genotype-bearing individuals within our 'summer' sub-cohort were at elevated risk of developing long-COVID, these findings point towards possible future use of IL-6 and/or rs2228145 genotype as biomarkers predictive of long-COVID risk, which may bring advantages regarding management and treatment of long-COVID.
Collapse
Affiliation(s)
- Katie Rees
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Rebecca Aicheler
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Lee Butcher
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Alan Dodd
- Clinical Biochemistry Department, Cwm Taf Morgannwg University Health Board, Merthyr Tydfil CF47 9DT, UK
| | - John Geen
- Clinical Biochemistry Department, Cwm Taf Morgannwg University Health Board, Merthyr Tydfil CF47 9DT, UK
| | - Ceri Lynch
- Intensive Care Unit, Cwm Taf Morgannwg University Health Board, Ynysmaerdy, Pontyclun, CF72 8XR, UK
| | - Isabel Massey
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Keith Morris
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Brian Tennant
- Clinical Biochemistry Department, Cwm Taf Morgannwg University Health Board, Merthyr Tydfil CF47 9DT, UK
| | - Richard Webb
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK.
| |
Collapse
|
2
|
Li X, Li Y, Zhang B, Wang J, Yang Y, Du Y. Plasma cytokine levels and PCOS risk: Mendelian randomization analysis reveals IL6R as a preventive factor. J Ovarian Res 2025; 18:69. [PMID: 40181376 PMCID: PMC11966818 DOI: 10.1186/s13048-025-01647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/15/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome is a prevalent gynecological condition affecting primarily women of childbearing age. It is characterized by elevated androgen levels, ovulatory dysfunction, and morphological abnormalities. Despite extensive research from various perspectives, the etiology and pathogenesis of PCOS remain unclear. While controversial, many believe that individuals with PCOS exhibit a chronic low-grade inflammatory state. Cytokines play diverse roles in the initiation and progression of inflammation, contributing to this inflammatory milieu. Therefore, the aim of this study was to utilize publicly available genome-wide association study data to explore the potential causal relationship between cytokines and PCOS. METHODS To accurately investigate the causal relationship between cytokines and PCOS, we initially defined cytokines using the GeneCrad and then identified cytokines in two independent large-scale plasma proteins. Subsequently, we employed a two-sample Mendelian randomization analysis framework. A series of quality control procedures were implemented to select eligible instrumental variables closely associated with the exposure. MR analysis was conducted using genome-wide association studies of PCOS in two independent European ancestry groups. Cochran, s Q test, MR-Egger and intercept test were employed to assess heterogeneity and pleiotropy in PCOS. Co-localization analysis, summary-data-based Mendelian randomization analysis, and HEIDI testing were utilized to further corroborate the relationship between positive findings and PCOS. Finally, systematical Mendelian randomization analysis between healthy lifestyle factors and PCOS-related proteins was conducted to identify which proteins could act as interventional targets by lifestyle changes. RESULTS In our investigation, we performed Mendelian randomization analysis on 33 cytokines in relation to PCOS using data from the deCODE and the Fenland. Our findings revealed that the plasma level of IL6R emerges as a notable protective factor against PCOS, exhibiting a substantial effect size. Moreover, we identified CCL22 as a significant risk factor for PCOS, a finding that was similarly validated and supported by independent cohorts. CONCLUSION Our Mendelian randomization analysis, leveraging genome-wide association study data from a sizable population cohort, unequivocally delineated a causal relationship between IL6R and PCOS. These results underscore the involvement of cytokines in the pathogenesis of PCOS and highlight their potential as promising therapeutic targets for addressing this intricate disease.
Collapse
Affiliation(s)
- Xiaopei Li
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yan Li
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bumei Zhang
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jianmei Wang
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Yang Yang
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China.
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China.
| | - Yongrui Du
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
3
|
Li C, Zhang C, Li X. Clonal hematopoiesis of indeterminate potential: contribution to disease and promising interventions. Mol Cell Biochem 2025:10.1007/s11010-025-05261-8. [PMID: 40140229 DOI: 10.1007/s11010-025-05261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/16/2025] [Indexed: 03/28/2025]
Abstract
In clonal hematopoiesis of indeterminate potential (CHIP), subpopulations of blood cells carrying somatic mutations expand as the individual ages, and this expansion may elevate risk of blood cancers as well as cardiovascular disease. Individuals at higher risk of CHIP and therefore of CHIP-associated disease can be identified through mutational profiling, and the apparently central role of inflammation in CHIP-associated disease has emerged as a potential therapeutic target. While CHIP is often associated with negative health outcomes, emerging evidence suggests that some CHIP-related mutations may also exert beneficial effects, indicating a more complex role in human health. This review examines current understanding of the epidemiology and clinical significance of CHIP and the role of inflammation in driving its association with disease risk. It explores the mechanisms linking CHIP to inflammation and risk of cardiovascular and other diseases, as well as the potential of personalizing therapies against those diseases for individuals with CHIP.
Collapse
Affiliation(s)
- Chongjie Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- School of Pharmacy, Southwest Medical University, LuZhou, 646000, Sichuan, People's Republic of China
| | - Chunxiang Zhang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- School of Pharmacy, Southwest Medical University, LuZhou, 646000, Sichuan, People's Republic of China.
| | - Xiuying Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- School of Pharmacy, Southwest Medical University, LuZhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
4
|
Blagov AV, Churov AV, Starodubtseva IA, Kovyanova TI, Pecherina TB, Sukhorukov VN. Cytokines are the Basis of the Development and Suppression of Inflammation in Atherosclerosis. Rev Cardiovasc Med 2025; 26:26421. [PMID: 40160593 PMCID: PMC11951496 DOI: 10.31083/rcm26421] [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: 09/03/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 04/02/2025] Open
Abstract
Cardiovascular diseases continue to be the primary cause of mortality in industrialised countries, and atherosclerosis plays a role in their development. A persistent inflammatory condition affecting big and medium-sized arteries is known as atherosclerosis. It is brought on by dyslipidemia and is facilitated by the immune system's innate and adaptive components. At every stage of the progression of atherosclerosis, inflammation plays a crucial role. It has been demonstrated that soluble factors, or cytokines, activate cells involved in the pathophysiology of atherosclerosis and have a significant impact on disease progression. Anti-inflammatory cytokines (such as interleukin (IL)-5 and IL-13) mitigate atherosclerosis, whereas pro-inflammatory cytokines (such as IL-1, IL-6) quicken the disease's course. Of interest is the fact that a number of cytokines can exhibit both atherogenic and atheroprotective properties, which is the topic of study and discussion in this review. This review provides a comparative analysis of the functions of the main cytokines involved in the pathogenesis of atherosclerosis. Their functional relationships with each other are also shown. In addition, potential therapeutic strategies targeting these cytokines for the treatment of atherosclerosis are proposed, with an emphasis on recent clinical research in this area.
Collapse
Affiliation(s)
| | - Alexey V. Churov
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Institute on Aging Research, Russian Gerontology Clinical Research Centre, Pirogov Russian National Research Medical University, 129226 Moscow, Russia
| | - Irina A. Starodubtseva
- Department of Polyclinic Therapy, NN Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Tatiana I. Kovyanova
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Institute for Atherosclerosis Research, 121609 Moscow, Russia
| | - Tamara B. Pecherina
- Laboratory for Cardiac Fibrosis, Research Institute for Complex Issues of Cardiovascular Diseases, 650002 Kemerovo, Russia
| | - Vasily N. Sukhorukov
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Institute of Experimental Cardiology, Russian Medical Research Center of Cardiology, 121552 Moscow, Russia
| |
Collapse
|
5
|
Zorc R, Redmond C, Sylvester M, Maclean M, Yamamoto de Almeida L, Quinn KA, Tomelleri A, Campochiaro C, Dagna L, Gutierrez-Rodrigues F, Wells KV, Rankin C, Hait SH, Palmer C, Corty R, Bick A, Lambert K, Buckner JH, O'Shea JJ, Park JK, Gadina M, Grayson PC. A coding single nucleotide polymorphism in the interleukin-6 receptor enhances IL-6 signalling in CD4 T cells and predicts treatment response to tocilizumab in giant cell arteritis. Ann Rheum Dis 2025:S0003-4967(25)00203-1. [PMID: 40000263 DOI: 10.1016/j.ard.2025.01.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/04/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025]
Abstract
OBJECTIVES The study objective was to determine if a common single nucleotide polymorphism in the interleukin 6 (IL-6) receptor (rs2228145, p.Asp358Ala) predicted treatment response to tocilizumab in giant cell arteritis (GCA). METHODS Genetic sequencing of the rs2228145 locus was performed in 2 independent cohorts of patients with GCA. Peripheral blood mononuclear cells (PBMCs) from patients and controls were evaluated for expression of the interleukin 6 receptor (IL-6R) and its coreceptor, gp130, using flow cytometry. The same PBMCs were stimulated with IL-6 and evaluated for downstream targets of IL-6: STAT3 phosphorylation (pSTAT3) and IL-17A expression. RESULTS In total, 100 patients with GCA were included (derivation cohort n = 58; validation cohort n = 42). The rs2228145 variant predicted tocilizumab response in each cohort. In the derivation cohort, a gene dose-dependent response was observed with a 36% response rate in the homozygous patients and 95% response rate in patients without the variant (P = .003). In the validation cohort, tocilizumab response rates were 50% for homozygotes and 85% for patients without the variant (P = .04). pSTAT3 levels were significantly increased in response to IL-6 stimulation in a gene dose-dependent manner in CD4 T cells from patients with GCA but not controls. CD4 T cells from patients with GCA had significantly higher membrane expression of gp130 than healthy controls, and response to IL-6 correlated with gp130 expression. IL-17 producing CD4 T cells were increased in a gene dose-dependent response to IL-6 (P < .01). CONCLUSIONS The rs2228145 variant is associated with decreased treatment response to tocilizumab and worse outcomes in GCA by enhancing CD4 T cell response to IL-6.
Collapse
Affiliation(s)
- Robert Zorc
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Redmond
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - McKella Sylvester
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mary Maclean
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luciana Yamamoto de Almeida
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kaitlin A Quinn
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alessandro Tomelleri
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Kristina V Wells
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cameron Rankin
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sabrina Helmold Hait
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chloe Palmer
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert Corty
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Alexander Bick
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Kathi Lambert
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Jane H Buckner
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - John J O'Shea
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jin Kyun Park
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA; Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Massimo Gadina
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter C Grayson
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Chen P, Hou S, Pang X, Li L, Wei W. A Proteomic Study Based on Home Quarantine Model Identifies NQO1 and Inflammation Pathways Involved in Adenoid Hypertrophy. J Inflamm Res 2025; 18:723-735. [PMID: 39845023 PMCID: PMC11750945 DOI: 10.2147/jir.s492921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
Background Adenoid hypertrophy is a common disorder of childhood, and has an unclear pathogenesis. At the beginning of the COVID-19 pandemic, there was a significant reduction in the incidence of adenoid hypertrophy in children under long-term home quarantine, providing a rare research model to explore the pathogenesis and treatment targets of adenoidal hypertrophy in children. Methodology Before and during the home quarantine period, adenoids that underwent surgery were detected using label-free proteomics. Differences in protein expression were analyzed using Gene Ontology, the Kyoto Encyclopedia of Genes and Genomes, Gene Set Enrichment Analysis, Protein-protein interaction, and immunohistochemistry analysis. Results Long-term home quarantine had a profound impact on the proteomics of pediatric adenoids, with up-regulated and down-regulated proteins of 28 and 92 downregulated proteins, respectively. Functional enrichment analysis showed that the differentially expressed proteins were mainly enriched in pathways such as leukocyte activation, inflammatory response, IL-1 production, Th17 cell differentiation, and IL-17 signaling. In the home quarantine group, inflammation-related proteins (TNF-α, IL-6), CD36, and S100A2, were considerably reduced, whereas NQO1 levels increased significantly, potentially alleviating adenoid hypertrophy. NQO1, CD36, NDUFS8, and NDUFAF2 exhibited strong interactions. Conclusion This study identified some candidate differential proteins, such as NQO1, CD36, S100A2, and the inflammation pathways involved in adenoid hypertrophy in preschool children.
Collapse
Affiliation(s)
- Penghui Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Shule Hou
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiuhong Pang
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, People’s Republic of China
| | - Lei Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Wei Wei
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
7
|
Pershad Y, Uddin MM, Xue L, Haessler J, Collins JM, Mack TM, Glick E, Glaser V, Zhao K, Jaiswal S, Manson JE, Pandey U, Desai P, Natarajan P, Honigberg MC, Kooperberg C, Whitsel EA, Kitzman JO, Bick AG, Reiner AP. Correlates and Consequences of Clonal Hematopoiesis Expansion Rate: A 15-Year Longitudinal Study of 6,986 Women. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.12.25320422. [PMID: 39867366 PMCID: PMC11759600 DOI: 10.1101/2025.01.12.25320422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is associated with increased mortality and malignancy risk, yet the determinants of clonal expansion remain poorly understood. We performed sequencing at >4,000x depth of coverage for CHIP mutations in 6,986 postmenopausal women from the Women's Health Initiative at two timepoints approximately 15 years apart. Among 3,685 mutations detected at baseline (VAF ≥ 0.5%), 50% progressed to CHIP (VAF ≥ 2%) at follow-up. We confirmed that clonal expansion is highly dependent on initial clone size and CHIP driver gene, with SF3B1 and JAK2 mutations exhibiting the fastest growth rate. We identified germline variants in TERT , IL6R , TCL1A , and MSI2 that modulate clonal expansion rate. Measured baseline leukocyte telomere length showed differential effects on incident CHIP risk, with shorter baseline leukocyte telomere length predisposing to incident PPM1D mutations and longer baseline leukocyte telomere length favoring incident DNMT3A mutations. We discovered that the IL6R missense variant p.Asp358Ala specifically impairs TET2 clonal expansion, supported by direct measurements of soluble interleukin-6 receptor and interleukin-6. Faster clonal growth rate was associated with increased risk of cytopenia, leukemia, and all-cause mortality. Notably, CHIP clonal expansion rate mediated 34.4% and 43.7% of the Clonal Hematopoiesis Risk Score's predictive value for leukemia and all-cause mortality, respectively. These findings reveal key biological determinants of CHIP progression and suggest that incorporating growth rate measurements could enhance risk stratification.
Collapse
|
8
|
Chuleerarux N, Makkoukdji N, Satnarine T, Kuhn JE, Nopsopon T, Valyasevi P, Schmidt FB, Kleiner G, Gans M. Inborn Errors of Immunity Presenting with Early-Onset Severe Atopy. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:62. [PMID: 39859044 PMCID: PMC11767231 DOI: 10.3390/medicina61010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025]
Abstract
Inborn errors of immunity (IEIs), also known as primary immunodeficiencies, are a group of genetic disorders affecting the development and function of the immune system. While IEIs traditionally present with recurrent infections, an increasing number of cases manifest with early-onset severe atopy, including atopic dermatitis, food allergies, asthma, and allergic rhinitis-features that are often overlooked. This can lead to delayed diagnosis and treatment, which is crucial for IEI patients due to the risk of severe infections. We conducted a literature search and reviewed all IEIs that can present with early-onset severe atopy. The hallmark features of these disorders often include early-onset, persistent, and severe atopic dermatitis, food allergies, and recurrent episodes of asthma, which may be refractory to treatments. Additionally, we discuss the importance of recognizing such severe atopy as a potential indicator of an underlying immune deficiency, particularly when accompanied by unusual infections, growth failure, or autoimmunity. This review aims to raise awareness of this association and emphasize the need for early diagnosis and genetic testing in patients with atypical or treatment-resistant allergic diseases, allowing for more timely diagnosis of underlying immunodeficiencies and appropriate treatments.
Collapse
Affiliation(s)
- Nipat Chuleerarux
- Department of Internal Medicine, Jackson Memorial Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Nadia Makkoukdji
- Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Travis Satnarine
- Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jessica Elise Kuhn
- Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Tanawin Nopsopon
- Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Peerada Valyasevi
- Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Fernanda Bellodi Schmidt
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Gary Kleiner
- Division of Allergy/Immunology, Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Melissa Gans
- Division of Allergy/Immunology, Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
9
|
Lin L, Li F, Fang H, Zhou P, Shan J, Xie K, Jin B, Zhu H, Jin X, Du L, Yang P. Associations of IL6R and IL10 Gene Polymorphisms with Susceptibility to Ankylosing Spondylitis with or without Acute Anterior Uveitis. Ocul Immunol Inflamm 2025; 33:2-9. [PMID: 38346238 DOI: 10.1080/09273948.2024.2309279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/08/2023] [Accepted: 01/17/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND This research aims to explore the associations between ten candidate single nucleotide polymorphisms (SNPs) on Interleukin-6 receptor (IL6R) and Interleukin-10 (IL10) genes and ankylosing spondylitis (AS) patients with or without acute anterior uveitis (AAU). METHODS This study involved a case-control approach that examined 354 cases with AS and AAU, 377 AS cases without AAU, and 918 healthy controls. Genotyping of ten SNPs of IL10 and IL6R genes was performed using iPLEX Gold genotyping method. The allele and genotype frequencies of cases and healthy individuals were contrasted using the chi-square test. The IL10 mRNA level in various IL10 genotypes was tested using real-time PCR. RESULTS Two loci associated with AS with AAU were identified: IL10//rs3790622 (OR = 0.664; 95%CI = 0.503-0.878; Pc = 0.038); IL10//rs3021094 (OR = 1.365; 95%CI = 1.110-1.679; Pc = 0.032). The other eight loci located on IL10 and IL6R did not show significant associations with the diseases. Additionally, as shown by functional experiments, there was no correlation between the mRNA expression of IL10 and various genotypes. CONCLUSION Our study suggests that the IL10 gene contributes to the susceptibility of the Chinese population to AS with AAU.
Collapse
Affiliation(s)
- Li Lin
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Fuzhen Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Haixin Fang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, P.R. China
| | - Pengyi Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Jiankang Shan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, P.R. China
| | - Kunpeng Xie
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Bo Jin
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Haiyan Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xuemin Jin
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Liping Du
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| |
Collapse
|
10
|
Hamilton F, Schurz H, Yates TA, Gilchrist JJ, Möller M, Naranbhai V, Ghazal P, Timpson NJ, Parks T, Pollara G. Altered IL-6 signalling and risk of tuberculosis: a multi-ancestry mendelian randomisation study. THE LANCET. MICROBE 2025; 6:100922. [PMID: 39579785 DOI: 10.1016/s2666-5247(24)00162-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND The role of IL-6 responses in human tuberculosis risk is unknown. IL-6 signalling inhibitors, such as tocilizumab, are thought to increase the risk of progression to tuberculosis, and screening for latent Mycobacterium tuberculosis infection before using these drugs is widely recommended. We used single nucleotide polymorphisms (SNPs) in and near the IL-6 receptor gene (IL6R), including the non-synonymous variant, rs2228145, for which the C allele contributes to reduced classic (cis) IL-6 signalling activity, to test the hypothesis that altered IL-6 signalling is causally associated with the risk of developing tuberculosis. METHODS We performed a meta-analysis of genome-wide association studies (GWAS) published in English from database inception to Jan 1, 2024. GWAS were identified from the European Bioinformatics Institute, MRC Integrative Epidemiology Unit catalogues, and MEDLINE, selecting publicly available studies for which tuberculosis was an outcome and that included the IL6R rs2228145 SNP. Using each study's population-level summary statistics, effect estimates were extracted for each additional copy of the C allele of rs2228145. We used these estimates to perform multi-ancestry, two-sample mendelian randomisation analyses to estimate the causal effect of reduced IL-6 signalling on tuberculosis. Our primary analyses used rs2228145-C as a genetic instrument, weighted on C-reactive protein (CRP) reduction as a measure of the effect on IL-6 signalling. We also took an alternative, ancestry-specific, multiple SNP approach using IL-6 receptor plasma protein as an exposure. Additionally, we compared the effects of rs2228145 in tuberculosis with those in critical COVID-19, rheumatoid arthritis, Crohn's disease, and coronary artery disease using the summary statistics extracted from GWAS. FINDINGS 17 GWAS were included, collating data for 19 302 individuals with tuberculosis (cases) and 1 019 821 population controls across multiple ancestries. For each additional rs2228145-C allele, the odds of tuberculosis reduced (odds ratio [OR] 0·94 [95% CI 0·92-0·97]; p=6·8 × 10-6). Multi-ancestry mendelian randomisation analyses supported these findings, with decreased odds of tuberculosis associated with readouts of reduced IL-6 signalling (0·52 [0·39-0·69] for each natural log CRP decrease; p=6·8 × 10-6), with weak evidence of heterogeneity (I2=0·315; p=0·11). Ancestry-specific, multiple SNP mendelian randomisation using increase in IL-6 receptor plasma protein as an exposure revealed a similar reduced risk of tuberculosis (OR 0·94 [95% CI 0·93-0·96]; p=2·4 × 10-10). The protective effects on tuberculosis seen with rs2228145-C were similar in size and direction to those observed in critical COVID-19 (0·66 [0·50-0·86]), Crohn's disease (0·57 [0·44-0·74]), and rheumatoid arthritis (0·45 [0·36-0·58]), all of which benefit from the therapeutic effects of IL-6 antagonism. INTERPRETATION Our findings propose a causal relationship between reduced IL-6 signalling and lower risk of tuberculosis, akin to the effect seen in other IL-6 mediated diseases. This study suggests that IL-6 antagonists do not increase the risk of tuberculosis but rather should be investigated as therapeutic adjuncts in its treatment. FUNDING UK National Institute for Health and Care Research, Wellcome Trust, EU European Regional Development Fund, the Welsh Government, and UK Research and Innovation.
Collapse
Affiliation(s)
- Fergus Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
| | - Haiko Schurz
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Tom A Yates
- Division of Infection and Immunity, University College London, London, UK; Institute of Health Informatics, University College London, London, UK
| | - James J Gilchrist
- Centre for Human Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK
| | - Marlo Möller
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vivek Naranbhai
- Centre for Human Genetics, University of Oxford, Oxford, UK; Massachusetts General Hospital, Boston, USA; Dana-Farber Cancer Institute, Boston, USA; Centre for the AIDS Programme of Research in South Africa, Durban, South Africa; Harvard Medical School, Boston, USA
| | | | | | - Tom Parks
- Centre for Human Genetics, University of Oxford, Oxford, UK; Department of Infectious Diseases Imperial College London, London, UK
| | - Gabriele Pollara
- Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|
11
|
Sheets K, Baker JV. HIV and Inflamm-Aging: How Do We Reach the Summit of Healthy Aging? TOPICS IN ANTIVIRAL MEDICINE 2024; 32:589-596. [PMID: 39765238 PMCID: PMC11737810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
People with HIV (PWH) are living longer and experiencing a greater burden of morbidity from non-AIDS-defining conditions. Chronically treated HIV disease is associated with ongoing systemic inflammation that contributes to the development of chronic conditions (eg, cardiovascular disease) and geriatric syndromes (eg, frailty). Apart from HIV disease, a progressive increase in systemic inflammation is a characteristic feature of biologic aging, a process described as "inflammaging." Inflamm-aging is driven by persistent antigen stimulation and stress, leading to an immune profile characterized by elevated levels of blood inflammatory markers and cellular activation and senescence. Chronic HIV disease is hypothesized to accentuate the immune profile of inflamm-aging, in part through viral persistence in lymphatic tissues, permanent injury impairing immune recovery, the presence of copathogens, gut dysbiosis and microbial translocation, and chromosomal and genetic alterations associated with immune activation. Few strategies exist for safe and effective modulation of systemic inflammation among older PWH. The strongest current evidence supports aggressive management of modifiable risk factors such as lipids, blood pressure, and levels of physical activity. Future inflamm-aging research should be directed toward advancing the implementation of proven approaches, such as physical activity, as well as studying novel mechanisms of, and treatments for, inflamm-aging among PWH.
Collapse
Affiliation(s)
- Kerry Sheets
- Hennepin Healthcare, Minneapolis, Minnesota, and University of Minnesota, Minneapolis
| | - Jason V. Baker
- Hennepin Healthcare, Minneapolis, Minnesota, and University of Minnesota, Minneapolis
| |
Collapse
|
12
|
Berger M, März W, Niessner A, Delgado G, Kleber M, Scharnagl H, Marx N, Schuett K. IL-6 and hsCRP predict cardiovascular mortality in patients with heart failure with preserved ejection fraction. ESC Heart Fail 2024; 11:3607-3615. [PMID: 39003598 PMCID: PMC11631318 DOI: 10.1002/ehf2.14959] [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: 02/12/2024] [Revised: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 07/15/2024] Open
Abstract
AIMS Inflammation accompanies heart failure (HF) and elevated levels of inflammatory biomarkers are linked to new onset of HF. However, whether the prognostic relevance of inflammatory biomarkers is different in HF with reduced (HFrEF) and preserved ejection fraction (HFpEF) is unclear. The aim of the current study is to explore the role of inflammation on the mortality risk in patients with HF. METHODS We analysed interleukin-6 and hsCRP levels by ELISA and immunonephelometry, respectively, in HFpEF and HFrEF patients referred for coronary angiography and assessed the prognostic value in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study. RESULTS HF was present in 1086 patients (N = 506 HFpEF; N = 580 HFrEF; mean age 65 ± 10 years; 28% female). Increasing IL-6 levels were significantly associated with increased CV mortality in HFpEF [1.5 (95% CI: 1.1-2.2), P = 0.018] but not HFrEF [HR 1.3 (95% CI: 1.0-1.7), P = 0.06] patients. High-sensitive CRP followed a similar pattern but failed to reach statistical significance after full-adjustment (HFpEF: HR 1.4 95%C I: 1.0-2.0; P = 0.065; HFrEF HR: 1.0 95% CI: 0.7-1.3; P = 0.800). Interaction analysis in patients stratified by IL-6 and N terminal pro brain natriuretic peptide (NT-proBNP) above and below the median revealed a stepwise increase in CV-mortality in HFpEF (P = 0.036) but not HFrEF patients (P = 0.220). To investigate the relationship between IL-6 and NT-proBNP, we assessed the genetic IL6-Receptor variant p.Asp358Ala (rs2228145) which is linked to impaired IL-6 receptor signalling. Homozygous carriers with HFpEF but not HFrEF exhibited significantly lower NT-pro-BNP levels compared with wildtype carriers (HFpEF 779 pg/mL ± 787 vs. 1180 pg/ mL ± 1532; P = 0.008; HFrEF 2289 pg/ mL ± 3439 vs. 2326 pg/ mL ± 3386; P = 0.94), raising the hypothesis that IL-6 signalling may play a pathophysiological role in HFpEF. CONCLUSIONS These data suggest a predictive value of elevated IL-6 for CV-mortality in HFpEF but not in HFrEF patients.
Collapse
Affiliation(s)
- Martin Berger
- Department of Internal Medicine I; University Hospital AachenRWTH UniversityAachenGermany
| | - Winfried März
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology) Mannheim Medical FacultyUniversity of HeidelbergMannheimGermany
- Synlab AcademySynlab Holding DeutschlandMannheimGermany
| | - Alexander Niessner
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaViennaAustria
| | - Graciela Delgado
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology) Mannheim Medical FacultyUniversity of HeidelbergMannheimGermany
| | - Marcus Kleber
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology) Mannheim Medical FacultyUniversity of HeidelbergMannheimGermany
- SYNLAB MVZ Humangenetik MannheimMannheimGermany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University of GrazGrazAustria
| | - Nikolaus Marx
- Department of Internal Medicine I; University Hospital AachenRWTH UniversityAachenGermany
| | - Katharina Schuett
- Department of Internal Medicine I; University Hospital AachenRWTH UniversityAachenGermany
| |
Collapse
|
13
|
Couch ACM, Brown AM, Raimundo C, Solomon S, Taylor M, Sichlinger L, Matuleviciute R, Srivastava DP, Vernon AC. Transcriptional and cellular response of hiPSC-derived microglia-neural progenitor co-cultures exposed to IL-6. Brain Behav Immun 2024; 122:27-43. [PMID: 39098436 DOI: 10.1016/j.bbi.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/12/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
Elevated interleukin (IL-)6 levels during prenatal development have been linked to increased risk for neurodevelopmental disorders (NDD) in the offspring, but the mechanism remains unclear. Human-induced pluripotent stem cell (hiPSC) models offer a valuable tool to study the effects of IL-6 on features relevant for human neurodevelopment in vitro. We previously reported that hiPSC-derived microglia-like cells (MGLs) respond to IL-6, but neural progenitor cells (NPCs) in monoculture do not. Therefore, we investigated whether co-culturing hiPSC-derived MGLs with NPCs would trigger a cellular response to IL-6 stimulation via secreted factors from the MGLs. Using N=4 donor lines without psychiatric diagnosis, we first confirmed that NPCs can respond to IL-6 through trans-signalling when recombinant IL-6Ra is present, and that this response is dose-dependent. MGLs secreted soluble IL-6R, but at lower levels than found in vivo and below that needed to activate trans-signalling in NPCs. Whilst transcriptomic and secretome analysis confirmed that MGLs undergo substantial transcriptomic changes after IL-6 exposure and subsequently secrete a cytokine milieu, NPCs in co-culture with MGLs exhibited a minimal transcriptional response. Furthermore, there were no significant cell fate-acquisition changes when differentiated into post-mitotic cultures, nor alterations in synaptic densities in mature neurons. These findings highlight the need to investigate if trans-IL-6 signalling to NPCs is a relevant disease mechanism linking prenatal IL-6 exposure to increased risk for psychiatric disorders. Moreover, our findings underscore the importance of establishing more complex in vitro human models with diverse cell types, which may show cell-specific responses to microglia-released cytokines to fully understand how IL-6 exposure may influence human neurodevelopment.
Collapse
Affiliation(s)
- Amalie C M Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| | - Amelia M Brown
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Catarina Raimundo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Shiden Solomon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Morgan Taylor
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Laura Sichlinger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Rugile Matuleviciute
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
14
|
Liu L, Zhu L, Monteiro-Martins S, Griffin A, Vlahos LJ, Fujita M, Berrouet C, Zanoni F, Marasa M, Zhang JY, Zhou XJ, Caliskan Y, Akchurin O, Al-Akash S, Jankauskiene A, Bodria M, Chishti A, Esposito C, Esposito V, Claes D, Tesar V, Davis TK, Samsonov D, Kaminska D, Hryszko T, Zaza G, Flynn JT, Iorember F, Lugani F, Rizk D, Julian BA, Hidalgo G, Kallash M, Biancone L, Amoroso A, Bono L, Mani LY, Vogt B, Lin F, Sreedharan R, Weng P, Ranch D, Xiao N, Quiroga A, Matar RB, Rheault MN, Wenderfer S, Selewski D, Lundberg S, Silva C, Mason S, Mahan JD, Vasylyeva TL, Mucha K, Foroncewicz B, Pączek L, Florczak M, Olszewska M, Gradzińska A, Szczepańska M, Machura E, Badeński A, Krakowczyk H, Sikora P, Kwella N, Miklaszewska M, Drożdż D, Zaniew M, Pawlaczyk K, SiniewiczLuzeńczyk K, Bomback AS, Appel GB, Izzi C, Scolari F, Materna-Kiryluk A, Mizerska-Wasiak M, Berthelot L, Pillebout E, Monteiro RC, Novak J, Green TJ, Smoyer WE, Hastings MC, Wyatt RJ, Nelson R, Martin J, González-Gay MA, De Jager PL, Köttgen A, Califano A, Gharavi AG, Zhang H, Kiryluk K. Genome-wide studies define new genetic mechanisms of IgA vasculitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.10.24315041. [PMID: 39417133 PMCID: PMC11482997 DOI: 10.1101/2024.10.10.24315041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
IgA vasculitis (IgAV) is a pediatric disease with skin and systemic manifestations. Here, we conducted genome, transcriptome, and proteome-wide association studies in 2,170 IgAV cases and 5,928 controls, generated IgAV-specific maps of gene expression and splicing from blood of 255 pediatric cases, and reconstructed myeloid-specific regulatory networks to define disease master regulators modulated by the newly identified disease driver genes. We observed significant association at the HLA-DRB1 (OR=1.55, P=1.1×10-25) and fine-mapped specific amino-acid risk substitutions in DRβ1. We discovered two novel non-HLA loci: FCAR (OR=1.51, P=1.0×10-20) encoding a myeloid IgA receptor FcαR, and INPP5D (OR=1.34, P=2.2×10-9) encoding a known inhibitor of FcαR signaling. The FCAR risk locus co-localized with a cis-eQTL increasing FCAR expression; the risk alleles disrupted a PRDM1 binding motif within a myeloid enhancer of FCAR. Another risk locus was associated with a higher genetically predicted levels of plasma IL6R. The IL6R risk haplotype carried a missense variant contributing to accelerated cleavage of IL6R into a soluble form. Using systems biology approaches, we prioritized IgAV master regulators co-modulated by FCAR, INPP5D and IL6R in myeloid cells. We additionally identified 21 shared loci in a cross-phenotype analysis of IgAV with IgA nephropathy, including novel loci PAID4, WLS, and ANKRD55.
Collapse
Affiliation(s)
- Lili Liu
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Li Zhu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Sara Monteiro-Martins
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aaron Griffin
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Lukas J. Vlahos
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Masashi Fujita
- Division of Neuroimmunology, Department of Neurology, Columbia University, New York, NY, USA
| | - Cecilia Berrouet
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Francesca Zanoni
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Maddalena Marasa
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Jun Y. Zhang
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Xu-jie Zhou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Yasar Caliskan
- Division of Nephrology, Saint Louis University, Saint Louis, MO, USA
| | | | | | | | - Monica Bodria
- MONICA BODRIA, MD, PHD, Primary Care Unit, Ausl Parma, south east district, Parma, Italy
| | - Aftab Chishti
- Division of Pediatric Nephrology, University of Kentucky, Kentucky Children’s Hospital, Lexington, KY, USA
| | - Ciro Esposito
- Istituti Clinico Scientifici Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Vittoria Esposito
- Istituti Clinico Scientifici Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Donna Claes
- Cinncinnati Children’s Hospital, Cincinnati, OH, USA
| | - Vladimir Tesar
- Department of Nephrology, 1st School of Medicine, Charles University Prague, Czech Republic
| | | | - Dmitry Samsonov
- Maria Fareri Children’s Hospital (MCF), New York Medical College, New York, NY, USA
| | - Dorota Kaminska
- Department of Non-Procedural Clinical Sciences, Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Tomasz Hryszko
- 2nd Department of Nephrology, Hypertension and Internal Medicine, Medical University of Bialystok, Poland
| | - Gianluigi Zaza
- Renal, Dialysis and Transplant Unit, Department of Pharmacy, Health and Nutritional Sciences (DFSSN), University of Calabria
| | - Joseph T. Flynn
- Department of Pediatrics, University of Washington; and Division of Nephrology, Seattle Children’s Hospital
| | | | | | - Dana Rizk
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | - Luisa Bono
- Nephrology and Dialysis, A.R.N.A.S. Civico and Benfratellio, Palermo, Italy
| | - Laila-Yasmin Mani
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fangming Lin
- Division of Pediatric Nephrology, Department of Medicine, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | - Scott Wenderfer
- Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Dave Selewski
- Mott Children’s Hospital, University of Michigan, Ann Arbor, MI, USA
| | - Sigrid Lundberg
- Danderyd University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Cynthia Silva
- Connecticut Children’s Medical Center, Hartford, CT, USA
| | - Sherene Mason
- Connecticut Children’s Medical Center, Hartford, CT, USA
| | | | | | - Krzysztof Mucha
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Bartosz Foroncewicz
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Pączek
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Michał Florczak
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | | | - Agnieszka Gradzińska
- Department of Dermatology and Pediatric Dermatology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Maria Szczepańska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Edyta Machura
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Andrzej Badeński
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Helena Krakowczyk
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Przemysław Sikora
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | - Norbert Kwella
- Department of Nephrology, Transplantology and Internal Diseases, University of Warmia and Mazury, Olsztyn, Poland
| | - Monika Miklaszewska
- Department of Pediatric Nephrology and Hypertension, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Marcin Zaniew
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland
| | - Krzysztof Pawlaczyk
- Department of Nephrology, Transplantology and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna SiniewiczLuzeńczyk
- Department of Paediatrics, Immunology and Nephrology, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | | | | | - Claudia Izzi
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | - Francesco Scolari
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | | | | | - Laureline Berthelot
- Nantes University, Inserm, CR2TI Center of Research on Translational Transplantation and Immunology, Nantes, France
| | - Evangeline Pillebout
- Center for Research on Inflammation, Paris Cité University, INSERM and CNRS, Paris, France
| | - Renato C. Monteiro
- Center for Research on Inflammation, Paris Cité University, INSERM and CNRS, Paris, France
| | - Jan Novak
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | - Robert J. Wyatt
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, Tennessee
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | | | - Javier Martin
- Institute of Parasitology and Biomedicine Lopez-Neyra, Spanish National Research Council (CSIC), Granada, Spain
| | - Miguel A. González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, Madrid, Spain
- Medicine and Psychiatry Department, University of Cantabria, Santander, Spain
| | - Philip L. De Jager
- Division of Neuroimmunology, Department of Neurology, Columbia University, New York, NY, USA
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Chan Zuckerberg Biohub New York, New York, NY, USA
| | - Ali G. Gharavi
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Krzysztof Kiryluk
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| |
Collapse
|
15
|
Wang SE, Viallon V, Lee M, Dimou N, Hamilton F, Biessy C, O'Mara T, Kyrgiou M, Crosbie EJ, Truong T, Severi G, Kaaks R, Fortner RT, Schulze MB, Bendinelli B, Sabina S, Tumino R, Sacerdote C, Panico S, Crous-Bou M, Sánchez MJ, Aizpurua A, Palacios DR, Guevara M, Travis RC, Tsilidis KK, Heath A, Yarmolinsky J, Rinaldi S, Gunter MJ, Dossus L. Circulating inflammatory and immune response proteins and endometrial cancer risk: a nested case-control study and Mendelian randomization analyses. EBioMedicine 2024; 108:105341. [PMID: 39278107 PMCID: PMC11418138 DOI: 10.1016/j.ebiom.2024.105341] [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: 05/25/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Inflammation and immune dysregulation are hypothesized contributors to endometrial carcinogenesis; however, the precise underlying mechanisms remain unclear. METHODS We measured pre-diagnostically 152 plasma protein biomarkers in 624 endometrial cancer case-control pairs nested within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Odds ratios (ORs) were estimated using conditional logistic regression, accounting for confounding and multiple comparisons. Proteins considered as associated with endometrial cancer risk were further tested in a two-sample Mendelian randomization (MR) analysis using summary data from the UK Biobank (n = 52,363) and the Endometrial Cancer Association Consortium (12,270 cases and 46,126 controls). FINDINGS In the EPIC nested case-control study, IL-6 [OR per NPX (doubling of concentration) = 1.28 (95% confidence interval (CI) 1.03-1.57)], HGF [1.48 (1.06-2.07)], PIK3AP1 [1.22 (1.00-1.50)] and CLEC4G [1.52 (1.00-2.32)] were positively associated; HSD11B1 [0.67 (0.49-0.91)], SCF [0.68 (0.49-0.94)], and CCL25 [0.80 (0.65-0.99)] were inversely associated with endometrial cancer risk; all estimates had multiple comparisons adjusted P-value > 0.05. In complementary MR analysis, IL-6 [OR per inverse-rank normalized NPX = 1.19 (95% CI 1.04-1.36)] and HSD11B1 [0.91 (0.84-0.99)] were associated with endometrial cancer risk. INTERPRETATION Altered IL-6 signalling and reduced glucocorticoid activity via HSD11B1 might play important roles in endometrial carcinogenesis. FUNDING Funding for IIG_FULL_2021_008 was obtained from Wereld Kanker Onderzoek Fonds (WKOF), as part of the World Cancer Research Fund International grant programme; Funding for INCA_15849 was obtained from Institut National du Cancer (INCa).
Collapse
Affiliation(s)
- Sabrina E Wang
- International Agency for Research on Cancer, World Health Organization, Lyon, France.
| | - Vivian Viallon
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Matthew Lee
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Niki Dimou
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Fergus Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Infection Science, North Bristol NHS Trust, Bristol, United Kingdom
| | - Carine Biessy
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Tracy O'Mara
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Maria Kyrgiou
- Department of Metabolism, Digestion & Reproduction - Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom; West London Gynaecological Cancer Centre, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Emma J Crosbie
- Gynaecological Oncology Research Group, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, United Kingdom; Department of Obstetrics and Gynaecology, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Therese Truong
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, CESP, Villejuif, France
| | - Gianluca Severi
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, CESP, Villejuif, France; Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Florence, Italy
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Renée Turzanski Fortner
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany; Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Benedetta Bendinelli
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Sieri Sabina
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Rosario Tumino
- Hyblean Association for Epidemiological Research, Aire Onlus, Ragusa, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza Hospital, University of Turin and CPO Piemonte, Turin, Italy
| | | | - Marta Crous-Bou
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO) - Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain; Instituto de Investigación Biosanitaria, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Amaia Aizpurua
- Sub Directorate for Public Health and Addictions of Gipuzkoa, Ministry of Health of the Basque Government, San Sebastian, Spain; Epidemiology of Chronic and Communicable Diseases Group, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Daniel Rodriguez Palacios
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; Department of Epidemiology, Murcia Regional Health Council-IMIB, Murcia, Spain
| | - Marcela Guevara
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; Instituto de Salud Pública y Laboral de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Ruth C Travis
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Konstantinos K Tsilidis
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom; Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Alicia Heath
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - James Yarmolinsky
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Sabina Rinaldi
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Marc J Gunter
- International Agency for Research on Cancer, World Health Organization, Lyon, France; Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Laure Dossus
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| |
Collapse
|
16
|
Wu M, Zhang X, Karunaratne S, Lee JH, Lampugnani ER, Selva KJ, Chung AW, Mueller SN, Chinnery HR, Downie LE. Intravital Imaging of the Human Cornea Reveals the Differential Effects of Season on Innate and Adaptive Immune Cell Morphodynamics. Ophthalmology 2024; 131:1185-1195. [PMID: 38703795 DOI: 10.1016/j.ophtha.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024] Open
Abstract
PURPOSE Defining how the in vivo immune status of peripheral tissues is shaped by the external environment has remained a technical challenge. We recently developed Functional in vivo confocal microscopy (Fun-IVCM) for dynamic, longitudinal imaging of corneal immune cells in living humans. This study investigated the effect of seasonal-driven environmental factors on the morphodynamic features of human corneal immune cell subsets. DESIGN Longitudinal, observational clinical study. PARTICIPANTS Sixteen healthy participants (aged 18-40 years) attended 2 visits in distinct seasons in Melbourne, Australia (Visit 1, November-December 2021 [spring-summer]; Visit 2, April-June 2022 [autumn-winter]). METHODS Environmental data were collected over each period. Participants underwent ocular surface examinations and corneal Fun-IVCM (Heidelberg Engineering). Corneal scans were acquired at 5.5 ± 1.5-minute intervals for up to 5 time points. Time-lapse Fun-IVCM videos were created to analyze corneal immune cells, comprising epithelial T cells and dendritic cells (DCs), and stromal macrophages. Tear cytokines were analyzed using a multiplex bead-based immunoassay. MAIN OUTCOME MEASURES Difference in the density, morphology, and dynamic parameters of corneal immune cell subsets over the study periods. RESULTS Visit 1 was characterized by higher temperature, lower humidity, and higher air particulate and pollen levels compared with Visit 2. Clinical ocular surface parameters and the density of immune cell subsets were similar across visits. At Visit 1 , corneal epithelial DCs were larger, with a lower dendrite probing speed (0.38 ± 0.21 vs. 0.68 ± 0.33 μm/min; P < 0.001) relative to Visit 2; stromal macrophages were more circular and had less dynamic activity (Visit 1, 7.2 ± 1.9 vs. Visit 2, 10.3 ± 3.7 dancing index; P < 0.001). Corneal T cell morphodynamics were unchanged across periods. Basal tear levels of interleukin 2 and CXCL10 were relatively lower during spring-summer. CONCLUSIONS This study identifies that the in vivo morphodynamics of innate corneal immune cells (DCs, macrophages) are modified by environmental factors, but such effects are not evident for adaptive immune cells (T cells). The cornea is a potential in vivo window to investigate season-dependent environmental influences on the human immune system. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Mengliang Wu
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia; Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Xinyuan Zhang
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
| | - Senuri Karunaratne
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
| | - Ji-Hyun Lee
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
| | - Edwin R Lampugnani
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia; Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kevin J Selva
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
| | - Laura E Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
| |
Collapse
|
17
|
Bashir U, Singh G, Bhatia A. Rheumatoid arthritis-recent advances in pathogenesis and the anti-inflammatory effect of plant-derived COX inhibitors. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5363-5385. [PMID: 38358467 DOI: 10.1007/s00210-024-02982-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024]
Abstract
The majority of people with autoimmune disorders, including those with rheumatoid arthritis, osteoarthritis, and tendonitis report pain, stiffness, and inflammation as major contributors to their worse quality of life in terms of overall health. Of all the available treatment options, COX inhibitors are the ones that are utilized most frequently to ease the symptoms. Various signaling cascades have been reported to be involved in the pathogenesis of rheumatoid arthritis which includes JAK/STAT, MAPK, and NF-kB signaling pathways, and several allopathic inhibitors (tofacitinib and baricitinib) have been reported to target the components of these cascades and have received approval for RA treatment. However, the prolonged use of these COX inhibitors and other allopathic drugs can pose serious health challenges due to their significant side effects. Therefore, searching for a more effective and side effect-free treatment for rheumatoid arthritis has unveiled phytochemicals as both productive and promising. Their therapeutic ability helps develop potent and safe drugs targeting immune-inflammatory diseases including RA. Various scientific databases were used for searching articles such as NCBI, SpringerLink, BioMed Central, ResearchGate, Google Scholar, Scopus, Nature, Wiley Online Library, and ScienceDirect. This review lists various phytochemicals and discusses their potential molecular targets in RA treatment, as demonstrated by various in vitro, in vivo (pre-clinical), and clinical studies. Several pre-clinical and clinical studies suggest that various phytochemicals can be an alternative promising intervention for attenuating and managing inflammation-associated pathogenesis of rheumatoid arthritis.
Collapse
Affiliation(s)
- Ubaid Bashir
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Gurjant Singh
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Astha Bhatia
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| |
Collapse
|
18
|
Zhao SS, Gill D. Genetically Proxied IL-6 Receptor Inhibition and Coronary Artery Disease Risk in a Japanese Population. Clin Ther 2024; 46:657-658. [PMID: 38821768 DOI: 10.1016/j.clinthera.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 06/02/2024]
Affiliation(s)
- Sizheng Steven Zhao
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Science, School of Biological Sciences, Faculty of Biological Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| |
Collapse
|
19
|
Kasher M, Freidin MB, Williams FMK, Livshits G. GlycA and CRP Are Genetically Correlated: Insight into the Genetic Architecture of Inflammageing. Biomolecules 2024; 14:563. [PMID: 38785970 PMCID: PMC11117775 DOI: 10.3390/biom14050563] [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: 04/07/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Inflammageing is a condition of perpetual low-grade inflammation induced by ageing. Inflammageing may be predicted by the C-reactive protein (CRP) or by a recently described biomarker which measures N-glycosylated side chains of the carbohydrate component of several acute-phase proteins known as GlycA. The objective of this study was to examine in depth the genetic relationships between CRP and GlycA as well as between each of them and other selected cytokines, which may shed light on the mechanisms of inflammageing. Using the Olink 96 Inflammation panel, data on inflammatory mediators for 1518 twins from the TwinsUK dataset were acquired. Summary statistics for genome-wide association studies for several cytokines as well as CRP and GlycA were collected from public sources. Extensive genetic correlation analyses, colocalization and genetic enrichment analyses were carried out to detect the shared genetic architecture between GlycA and CRP. Mendelian randomization was carried out to assess potential causal relationships. GlycA predicted examined cytokines with a magnitude twice as great as that of CRP. GlycA and CRP were significantly genetically correlated (Rg = 0.4397 ± 0.0854, p-value = 2.60 × 10-7). No evidence of a causal relationship between GlycA and CRP, or between these two biomarkers and the cytokines assessed was obtained. However, the aforementioned relationships were explained well by horizontal pleiotropy. Five exonic genetic variants annotated to five genes explain the shared genetic architecture observed between GlycA and CRP: IL6R, GCKR, MLXIPL, SERPINA1, and MAP1A. GlycA and CRP possess a shared genetic architecture, but the relationship between them appears to be modest, which may imply the promotion of differing inflammatory pathways. GlycA appears to be a more robust predictor of cytokines compared to CRP.
Collapse
Affiliation(s)
- Melody Kasher
- Department of Morphological Sciences, Adelson Medical School, Ariel University, Ariel 40700, Israel;
| | - Maxim B. Freidin
- Department of Biology, School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK;
| | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King’s College London, London SE1 7EH, UK;
| | - Gregory Livshits
- Department of Morphological Sciences, Adelson Medical School, Ariel University, Ariel 40700, Israel;
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King’s College London, London SE1 7EH, UK;
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | |
Collapse
|
20
|
Iakunchykova O, Pan M, Amlien IK, Roe JM, Walhovd KB, Fjell AM, Chen CH, Benros ME, Wang Y. Genetic evidence for the causal effects of C-reactive protein on self-reported habitual sleep duration. Brain Behav Immun Health 2024; 37:100754. [PMID: 38511149 PMCID: PMC10950822 DOI: 10.1016/j.bbih.2024.100754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/13/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
Inflammatory responses to acute stimuli are proposed to regulate sleep, but the relationship between chronic inflammation and habitual sleep duration is elusive. Here, we study this relation using genetically predicted level of chronic inflammation, indexed by CRP and IL6 signaling, and self-reported sleep duration. By Mendelian randomization analysis, we show that elevated CRP level within <10 mg/L has a homeostatic effect that facilitates maintaining 7-8 h sleep duration per day - making short-sleepers sleep longer (p = 2.42 × 10-2) and long-sleepers sleep shorter (1.87 × 10-7); but it is not associated with the overall sleep duration (p = 0.17). This homeostatic effect replicated in an independent CRP dataset. We observed causal effects of the soluble interleukin 6 receptor and gp130 on overall sleep duration (p = 1.62 × 10-8, p = 2.61 × 10-58, respectively), but these effects disappeared when CRP effects were accounted for in the model. Using polygenic score analysis, we found that the homeostatic effect of CRP on sleep duration stems primarily from the genetic variants within the CRP gene region: when genetic variants outside of this region were used to predict CRP levels, the opposite direction of effect was observed. In conclusion, we show that elevated CRP level may causally facilitate maintaining an optimal sleep duration that is beneficial to health, thus updating our current knowledge of immune regulation on sleep.
Collapse
Affiliation(s)
- Olena Iakunchykova
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway
| | - Mengyu Pan
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway
| | - Inge K. Amlien
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway
| | - James M. Roe
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway
| | - Kristine B. Walhovd
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Rikshospitalet, POB 4950, Nydalen, 0424, Oslo, Norway
| | - Anders M. Fjell
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Rikshospitalet, POB 4950, Nydalen, 0424, Oslo, Norway
| | - Chi-Hua Chen
- Department of Radiology, University of California in San Diego, Gilman Drive 9500, 92093, La Jolla, CA, USA
| | - Michael E. Benros
- Copenhagen Research Centre for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Gentofte Hospitalsvej 15, 2900, Hellerup, Denmark
| | - Yunpeng Wang
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway
| |
Collapse
|
21
|
Pierce GF, Fong S, Long BR, Kaczmarek R. Deciphering conundrums of adeno-associated virus liver-directed gene therapy: focus on hemophilia. J Thromb Haemost 2024; 22:1263-1289. [PMID: 38103734 DOI: 10.1016/j.jtha.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/07/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Adeno-associated virus gene therapy has been the subject of intensive investigation for monogenic disease gene addition therapy for more than 25 years, yet few therapies have been approved by regulatory agencies. Most have not progressed beyond phase 1/2 due to toxicity, lack of efficacy, or both. The liver is a natural target for adeno-associated virus since most serotypes have a high degree of tropism for hepatocytes due to cell surface receptors for the virus and the unique liver sinusoidal geometry facilitating high volumes of blood contact with hepatocyte cell surfaces. Recessive monogenic diseases such as hemophilia represent promising targets since the defective proteins are often synthesized in the liver and secreted into the circulation, making them easy to measure, and many do not require precise regulation. Yet, despite initiation of many disease-specific clinical trials, therapeutic windows are often nonexistent, resulting in excess toxicity and insufficient efficacy. Iterative progress built on these attempts is best illustrated by hemophilia, with the first regulatory approvals for factor IX and factor VIII gene therapies eventually achieved 25 years after the first gene therapy studies in humans. Although successful gene transfer may result in the production of sufficient transgenic protein to modify the disease, many emerging questions on durability, predictability, reliability, and variability of response have not been answered. The underlying biology accounting for these heterogeneous responses and the interplay between host and virus is the subject of intense investigation and the subject of this review.
Collapse
Affiliation(s)
- Glenn F Pierce
- World Federation of Hemophilia, Montreal, Quebec, Canada.
| | - Sylvia Fong
- BioMarin Pharmaceutical Inc, Research and Early Development, Novato, California, USA
| | - Brian R Long
- BioMarin Pharmaceutical Inc, Research and Early Development, Novato, California, USA
| | - Radoslaw Kaczmarek
- Department of Pediatrics, Indiana University School of Medicine, Wells Center for Pediatric Research, Indiana, USA; Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| |
Collapse
|
22
|
Wang X, Liu M, Nogues IE, Chen T, Xiong X, Bonzel CL, Zhang H, Hong C, Xia Y, Dahal K, Costa L, Cui J, Gaziano JM, Kim SC, Ho YL, Cho K, Cai T, Liao KP. Heterogeneous associations between interleukin-6 receptor variants and phenotypes across ancestries and implications for therapy. Sci Rep 2024; 14:8021. [PMID: 38580710 PMCID: PMC10997791 DOI: 10.1038/s41598-024-54063-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/08/2024] [Indexed: 04/07/2024] Open
Abstract
The Phenome-Wide Association Study (PheWAS) is increasingly used to broadly screen for potential treatment effects, e.g., IL6R variant as a proxy for IL6R antagonists. This approach offers an opportunity to address the limited power in clinical trials to study differential treatment effects across patient subgroups. However, limited methods exist to efficiently test for differences across subgroups in the thousands of multiple comparisons generated as part of a PheWAS. In this study, we developed an approach that maximizes the power to test for heterogeneous genotype-phenotype associations and applied this approach to an IL6R PheWAS among individuals of African (AFR) and European (EUR) ancestries. We identified 29 traits with differences in IL6R variant-phenotype associations, including a lower risk of type 2 diabetes in AFR (OR 0.96) vs EUR (OR 1.0, p-value for heterogeneity = 8.5 × 10-3), and higher white blood cell count (p-value for heterogeneity = 8.5 × 10-131). These data suggest a more salutary effect of IL6R blockade for T2D among individuals of AFR vs EUR ancestry and provide data to inform ongoing clinical trials targeting IL6 for an expanding number of conditions. Moreover, the method to test for heterogeneity of associations can be applied broadly to other large-scale genotype-phenotype screens in diverse populations.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Molei Liu
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | | | - Tony Chen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Xin Xiong
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Clara-Lea Bonzel
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Harrison Zhang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Chuan Hong
- Department of Biostatistics, Duke University, Durham, NC, USA
| | - Yin Xia
- Department of Statistics and Data Science, Fudan University, Shanghai, China
| | - Kumar Dahal
- Department of Biostatistics, Duke University, Durham, NC, USA
| | - Lauren Costa
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA
| | - Jing Cui
- Department of Biostatistics, Duke University, Durham, NC, USA
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Boston, MA, USA
| | - Seoyoung C Kim
- Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women's Hospital, Boston, MA, USA
| | - Yuk-Lam Ho
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Boston, MA, USA
| | - Tianxi Cai
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
| | - Katherine P Liao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA.
- Rheumatology Section, VA Boston Healthcare System, Boston, USA.
| |
Collapse
|
23
|
Prapiadou S, Živković L, Thorand B, George MJ, van der Laan SW, Malik R, Herder C, Koenig W, Ueland T, Kleveland O, Aukrust P, Gullestad L, Bernhagen J, Pasterkamp G, Peters A, Hingorani AD, Rosand J, Dichgans M, Anderson CD, Georgakis MK. Proteogenomic Data Integration Reveals CXCL10 as a Potentially Downstream Causal Mediator for IL-6 Signaling on Atherosclerosis. Circulation 2024; 149:669-683. [PMID: 38152968 PMCID: PMC10922752 DOI: 10.1161/circulationaha.123.064974] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 11/17/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Genetic and experimental studies support a causal involvement of IL-6 (interleukin-6) signaling in atheroprogression. Although trials targeting IL-6 signaling are underway, any benefits must be balanced against an impaired host immune response. Dissecting the mechanisms that mediate the effects of IL-6 signaling on atherosclerosis could offer insights about novel drug targets with more specific effects. METHODS Leveraging data from 522 681 individuals, we constructed a genetic instrument of 26 variants in the gene encoding the IL-6R (IL-6 receptor) that proxied for pharmacological IL-6R inhibition. Using Mendelian randomization, we assessed its effects on 3281 plasma proteins quantified with an aptamer-based assay in the INTERVAL cohort (n=3301). Using mediation Mendelian randomization, we explored proteomic mediators of the effects of genetically proxied IL-6 signaling on coronary artery disease, large artery atherosclerotic stroke, and peripheral artery disease. For significant mediators, we tested associations of their circulating levels with incident cardiovascular events in a population-based study (n=1704) and explored the histological, transcriptomic, and cellular phenotypes correlated with their expression levels in samples from human atherosclerotic lesions. RESULTS We found significant effects of genetically proxied IL-6 signaling on 70 circulating proteins involved in cytokine production/regulation and immune cell recruitment/differentiation, which correlated with the proteomic effects of pharmacological IL-6R inhibition in a clinical trial. Among the 70 significant proteins, genetically proxied circulating levels of CXCL10 (C-X-C motif chemokine ligand 10) were associated with risk of coronary artery disease, large artery atherosclerotic stroke, and peripheral artery disease, with up to 67% of the effects of genetically downregulated IL-6 signaling on these end points mediated by decreases in CXCL10. Higher midlife circulating CXCL10 levels were associated with a larger number of cardiovascular events over 20 years, whereas higher CXCL10 expression in human atherosclerotic lesions correlated with a larger lipid core and a transcriptomic profile reflecting immune cell infiltration, adaptive immune system activation, and cytokine signaling. CONCLUSIONS Integrating multiomics data, we found a proteomic signature of IL-6 signaling activation and mediators of its effects on cardiovascular disease. Our analyses suggest the interferon-γ-inducible chemokine CXCL10 to be a potentially causal mediator for atherosclerosis in 3 vascular compartments and, as such, could serve as a promising drug target for atheroprotection.
Collapse
Affiliation(s)
- Savvina Prapiadou
- University of Patras School of Medicine, Patras, Greece
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Luka Živković
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Marc J. George
- Department of Clinical Pharmacology, Division of Medicine, University College London, London, United Kingdom
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rainer Malik
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Wolfgang Koenig
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
- German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Thor Ueland
- Thrombosis Research Center (TREC), Division of internal medicine, University hospital of North Norway, Tromsø, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ola Kleveland
- Clinic of Cardiology, St Olavs Hospital, Trondheim, Norway
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars Gullestad
- Department of Cardiology Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig-Maximilians-University Munich, Munich, Germany
- Munich Heart Alliance, German Center for Cardiovascular Health (DZHK e.V., partner-site Munich), Munich, Germany
| | - Aroon D. Hingorani
- Department of Clinical Pharmacology, Division of Medicine, University College London, London, United Kingdom
- Centre for Translational Genomics, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Jonathan Rosand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Christopher D. Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Marios K. Georgakis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
24
|
Colvin A, Youssef S, Noh H, Wright J, Jumonville G, LaRow Brown K, Tatonetti NP, Milner JD, Weng C, Bordone LA, Petukhova L. Inborn Errors of Immunity Contribute to the Burden of Skin Disease and Create Opportunities for Improving the Practice of Dermatology. J Invest Dermatol 2024; 144:307-315.e1. [PMID: 37716649 DOI: 10.1016/j.jid.2023.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 09/18/2023]
Abstract
Opportunities to improve the clinical management of skin disease are being created by advances in genomic medicine. Large-scale sequencing increasingly challenges notions about single-gene disorders. It is now apparent that monogenic etiologies make appreciable contributions to the population burden of disease and that they are underrecognized in clinical practice. A genetic diagnosis informs on molecular pathology and may direct targeted treatments and tailored prevention strategies for patients and family members. It also generates knowledge about disease pathogenesis and management that is relevant to patients without rare pathogenic variants. Inborn errors of immunity are a large class of monogenic etiologies that have been well-studied and contribute to the population burden of inflammatory diseases. To further delineate the contributions of inborn errors of immunity to the pathogenesis of skin disease, we performed a set of analyses that identified 316 inborn errors of immunity associated with skin pathologies, including common skin diseases. These data suggest that clinical sequencing is underutilized in dermatology. We next use these data to derive a network that illuminates the molecular relationships of these disorders and suggests an underlying etiological organization to immune-mediated skin disease. Our results motivate the further development of a molecularly derived and data-driven reorganization of clinical diagnoses of skin disease.
Collapse
Affiliation(s)
- Annelise Colvin
- Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Soundos Youssef
- Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Heeju Noh
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Julia Wright
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Ghislaine Jumonville
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Kathleen LaRow Brown
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Nicholas P Tatonetti
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA; Department of Computational Biomedicine, Cedars-Sinai Medical Center, West Hollywood, California, USA; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Joshua D Milner
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Chunhua Weng
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Lindsey A Bordone
- Department of Dermatology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Lynn Petukhova
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA; Department of Dermatology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA.
| |
Collapse
|
25
|
Ebrat E, Hejazian SM, Ahmadian E, Vahed SZ, Mobasseri M, Ardalan M. The Possible Association of IL-6R Gene Polymorphisms in the Development of Diabetic Nephropathy. Curr Diabetes Rev 2024; 20:55-59. [PMID: 37855360 DOI: 10.2174/0115733998245369231009111221] [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: 03/16/2023] [Revised: 08/31/2023] [Accepted: 09/08/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a common complication of type 2 diabetes (T2D). Chronic inflammation and a combination of environmental and genetic factors are involved in the pathogenesis and development of DN. OBJECTIVE This case-control study aimed to determine the relationship between rs7529229 and rs2228145 polymorphisms of the IL-6R gene with the incidence of nephropathy among T2D patients. METHODS Fifty-six diabetic patients with nephropathy and 57 T2D patients without nephropathy were included based on inclusion criteria, along with 150 healthy individuals. RESULTS The frequencies of AC and CC genotype distributions of the rs2228145 polymorphism in DN patients were significantly higher than in healthy individuals (24.1 and 9.3% versus 10.7 and 6.7%, respectively, P= 0.02). Moreover, the frequency of allele C was higher in DN patients compared to healthy controls (21.30% versus 12%, P=0.025). However, genotype distribution and allele frequencies of the rs7529229 IL-6R polymorphism in DN patients were not statistically significant in comparison with diabetic patients and healthy individuals (P> 0.05). CONCLUSION The results showed that the allele and genotype distribution frequencies of rs2228145 IL-6R gene polymorphism in patients with DN were significantly higher than in healthy individuals. Therefore, the presence of this polymorphism may be involved in the development of diabetic nephropathy in this population.
Collapse
Affiliation(s)
- Ebrahim Ebrat
- Kidney Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyedeh Mina Hejazian
- Kidney Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Kidney Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Zununi Vahed
- Kidney Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Mobasseri
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Ardalan
- Kidney Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
Henriques P, Rosa A, Caldeira-Araújo H, Soares P, Vigário AM. Flying under the radar - impact and factors influencing asymptomatic DENV infections. Front Cell Infect Microbiol 2023; 13:1284651. [PMID: 38076464 PMCID: PMC10704250 DOI: 10.3389/fcimb.2023.1284651] [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: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
The clinical outcome of DENV and other Flaviviruses infections represents a spectrum of severity that ranges from mild manifestations to severe disease, which can ultimately lead to death. Nonetheless, most of these infections result in an asymptomatic outcome that may play an important role in the persistent circulation of these viruses. Also, although little is known about the mechanisms that lead to these asymptomatic infections, they are likely the result of a complex interplay between viral and host factors. Specific characteristics of the infecting viral strain, such as its replicating efficiency, coupled with host factors, like gene expression of key molecules involved in the immune response or in the protection against disease, are among crucial factors to study. This review revisits recent data on factors that may contribute to the asymptomatic outcome of the world's widespread DENV, highlighting the importance of silent infections in the transmission of this pathogen and the immune status of the host.
Collapse
Affiliation(s)
- Paulo Henriques
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Alexandra Rosa
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Helena Caldeira-Araújo
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- CQM-Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Pedro Soares
- Department of Biology, CBMA (Centre of Molecular and Environmental Biology), Braga, Portugal
- Department of Biology, Institute of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, Braga, Portugal
| | - Ana Margarida Vigário
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
27
|
Sainz L, Riera P, Moya P, Bernal S, Casademont J, Díaz-Torné C, Millán AM, Park HS, Lasa A, Corominas H. Impact of IL6R genetic variants on treatment efficacy and toxicity response to sarilumab in rheumatoid arthritis. Arthritis Res Ther 2023; 25:226. [PMID: 38001504 PMCID: PMC10668502 DOI: 10.1186/s13075-023-03209-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Sarilumab, an IL-6 receptor antagonist, is a first-line biologic disease-modifying anti-rheumatic drug for rheumatoid arthritis. The identification of genetic biomarkers as predictors of response to sarilumab could allow for a personalized treatment strategy to improve clinical outcomes. METHODS We conducted a retrospective cohort study of 62 patients treated with sarilumab to determine whether single-nucleotide polymorphisms (SNP) in the IL6R gene could predict efficacy and toxicity responses. Six SNPs previously described in the IL6R gene (rs12083537, rs11265618, rs4329505, rs2228145, rs4537545, and rs4845625) were genotyped in DNA samples obtained from these patients. Using parametric tests, we evaluated the association between these polymorphisms and clinicopathological features. Treatment response was assessed six months after treatment initiation. Satisfactory response was based on EULAR criteria. Low disease activity was determined according to DAS28 and CDAI and quantitative improvements in DAS28 and CDAI scores. RESULTS Three SNPs (rs4845625, rs4329505 and rs11265618) were significantly associated with response outcomes. All of the SNPs, except for rs12083537, had at least one significant association with dyslipidemia or hepatotoxicity. CONCLUSIONS These findings support the potential clinical value of SNPs, particularly rs4845625, as potentially useful biomarkers to predict response to sarilumab in patients with RA.
Collapse
Affiliation(s)
- Luis Sainz
- Rheumatology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Pau Riera
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.
- Pharmacy Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain.
- Biomedical Network Research Centre On Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.
| | - Patricia Moya
- Rheumatology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Sara Bernal
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Biomedical Network Research Centre On Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Genetics Department, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Jordi Casademont
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Internal Medicine Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Cesar Díaz-Torné
- Rheumatology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Ana Milena Millán
- Rheumatology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Hye Sang Park
- Rheumatology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Adriana Lasa
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Biomedical Network Research Centre On Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Genetics Department, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Hector Corominas
- Rheumatology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain.
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.
| |
Collapse
|
28
|
Rose-John S, Jenkins BJ, Garbers C, Moll JM, Scheller J. Targeting IL-6 trans-signalling: past, present and future prospects. Nat Rev Immunol 2023; 23:666-681. [PMID: 37069261 PMCID: PMC10108826 DOI: 10.1038/s41577-023-00856-y] [Citation(s) in RCA: 168] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 04/19/2023]
Abstract
Interleukin-6 (IL-6) is a key immunomodulatory cytokine that affects the pathogenesis of diverse diseases, including autoimmune diseases, chronic inflammatory conditions and cancer. Classical IL-6 signalling involves the binding of IL-6 to the membrane-bound IL-6 receptor α-subunit (hereafter termed 'mIL-6R') and glycoprotein 130 (gp130) signal-transducing subunit. By contrast, in IL-6 trans-signalling, complexes of IL-6 and the soluble form of IL-6 receptor (sIL-6R) signal via membrane-bound gp130. A third mode of IL-6 signalling - known as cluster signalling - involves preformed complexes of membrane-bound IL-6-mIL-6R on one cell activating gp130 subunits on target cells. Antibodies and small molecules have been developed that block all three forms of IL-6 signalling, but in the past decade, IL-6 trans-signalling has emerged as the predominant pathway by which IL-6 promotes disease pathogenesis. The first selective inhibitor of IL-6 trans-signalling, sgp130, has shown therapeutic potential in various preclinical models of disease and olamkicept, a sgp130Fc variant, had promising results in phase II clinical studies for inflammatory bowel disease. Technological developments have already led to next-generation sgp130 variants with increased affinity and selectivity towards IL-6 trans-signalling, along with indirect strategies to block IL-6 trans-signalling. Here, we summarize our current understanding of the biological outcomes of IL-6-mediated signalling and the potential for targeting this pathway in the clinic.
Collapse
Affiliation(s)
- Stefan Rose-John
- Biochemical Institute, Medical Faculty, Christian-Albrechts-University, Kiel, Germany
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC:I3), Otto-von-Guericke-University, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
29
|
Zhao JH, Stacey D, Eriksson N, Macdonald-Dunlop E, Hedman ÅK, Kalnapenkis A, Enroth S, Cozzetto D, Digby-Bell J, Marten J, Folkersen L, Herder C, Jonsson L, Bergen SE, Gieger C, Needham EJ, Surendran P, Paul DS, Polasek O, Thorand B, Grallert H, Roden M, Võsa U, Esko T, Hayward C, Johansson Å, Gyllensten U, Powell N, Hansson O, Mattsson-Carlgren N, Joshi PK, Danesh J, Padyukov L, Klareskog L, Landén M, Wilson JF, Siegbahn A, Wallentin L, Mälarstig A, Butterworth AS, Peters JE. Genetics of circulating inflammatory proteins identifies drivers of immune-mediated disease risk and therapeutic targets. Nat Immunol 2023; 24:1540-1551. [PMID: 37563310 PMCID: PMC10457199 DOI: 10.1038/s41590-023-01588-w] [Citation(s) in RCA: 272] [Impact Index Per Article: 136.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/13/2023] [Indexed: 08/12/2023]
Abstract
Circulating proteins have important functions in inflammation and a broad range of diseases. To identify genetic influences on inflammation-related proteins, we conducted a genome-wide protein quantitative trait locus (pQTL) study of 91 plasma proteins measured using the Olink Target platform in 14,824 participants. We identified 180 pQTLs (59 cis, 121 trans). Integration of pQTL data with eQTL and disease genome-wide association studies provided insight into pathogenesis, implicating lymphotoxin-α in multiple sclerosis. Using Mendelian randomization (MR) to assess causality in disease etiology, we identified both shared and distinct effects of specific proteins across immune-mediated diseases, including directionally discordant effects of CD40 on risk of rheumatoid arthritis versus multiple sclerosis and inflammatory bowel disease. MR implicated CXCL5 in the etiology of ulcerative colitis (UC) and we show elevated gut CXCL5 transcript expression in patients with UC. These results identify targets of existing drugs and provide a powerful resource to facilitate future drug target prioritization.
Collapse
Affiliation(s)
- Jing Hua Zhao
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - David Stacey
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Australian Centre for Precision Health, Unit of Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Erin Macdonald-Dunlop
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Åsa K Hedman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Development and Medical, Pfizer Worldwide Research, Stockholm, Sweden
| | - Anette Kalnapenkis
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Stefan Enroth
- Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, Uppsala, Sweden
| | - Domenico Cozzetto
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Jonathan Digby-Bell
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Jonathan Marten
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Munich-Neuherberg, Germany
| | - Lina Jonsson
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Sarah E Bergen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Elise J Needham
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
| | - Dirk S Paul
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Barbara Thorand
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Harald Grallert
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Munich-Neuherberg, Germany
| | - Urmo Võsa
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tonu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Åsa Johansson
- Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, Uppsala, Sweden
| | - Ulf Gyllensten
- Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, Uppsala, Sweden
| | - Nick Powell
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Leonid Padyukov
- Division of Rheumatology, Department of Medicine (Solna), Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars Klareskog
- Division of Rheumatology, Department of Medicine (Solna), Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Agneta Siegbahn
- Department of Medical Sciences and Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Department of Medical Sciences and Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Anders Mälarstig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Development and Medical, Pfizer Worldwide Research, Stockholm, Sweden
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, Hinxton, UK.
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK.
| | - James E Peters
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, Hinxton, UK.
- Department of Immunology and Inflammation, Imperial College London, London, UK.
| |
Collapse
|
30
|
Yu M, Li Y, Li B, Ge Q. Inflammatory biomarkers and delirium: a Mendelian randomization study. Front Aging Neurosci 2023; 15:1221272. [PMID: 37649721 PMCID: PMC10464607 DOI: 10.3389/fnagi.2023.1221272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/25/2023] [Indexed: 09/01/2023] Open
Abstract
Background The association between inflammatory biomarkers and individual delirium symptoms remains controversial in observational studies. We investigated the relationship between inflammatory biomarkers and the risk of developing delirium. Methods A bidirectional two-sample Mendelian randomization (MR) was performed. Genetic instruments associated with peripheral tumor necrosis factor-a (TNF-a) C-reactive protein (CRP), interleukin (IL)-1α, IL-1β, IL-2, IL-8, IL-6, soluble IL-6 receptor alpha (sIL-6Rα), and soluble gp130 were identified in three different large summary genome-wide association studies (GWAS) conducted in the European population. Summary-level statistics for delirium not induced by alcohol and other psychoactive substances were obtained from the FinnGen consortium (2,612 cases and 325,306 controls). The estimated causal effects were performed using instruments' variants at the genome-wide significant level (P < 5e-8 and P < 5e-6), applying a linkage disequilibrium clumping approach with a threshold of r2 < 0.001 for each of the exposures. Reverse causation was also performed. The inverse-variance weighted method (IVW), MR-Egger method, weighted median method, MR-Egger regression, and MR Pleiotropy RESidual Sum were used for MR analyses. Results At the genome-wide significant level (P < 5e-8, r2 < 0.001), genetically predicted sIL-6Rα was significantly associated with a decreased risk of delirium with less than three single-nucleotide polymorphisms (SNPs) in all three GWAS data sources (ORWaldratio = 0.89, 95% CI: 0.79-0.96, PWaldratio = 0.0016; ORIVW = 0.88, 95% CI: 0.79-0.97, PIVW = 0.008; ORIVW = 0.88, 95% CI: 0.80-0.96, PIVW = 0.004). The causal relationship between sIL-6Rα and delirium became non-significant when a more liberal threshold of P of < 5e-6 was applied (all PIVW > 0.05). At the two genome-wide significance levels (P < 5e-8 and P < 5e-6), we found no evidence for the causal effects of peripheral TNF-α, CRP, IL-1α, IL-1β, IL-2, IL-6, IL-8, and soluble gp130 on delirium (all P > 0.05). The MR-Egger intercept and MR-PRESSO results indicated that no SNP had possible pleiotropy (all P > 0.05). Regarding the reverse, no evidence for an effect of delirium on these inflammatory biomarkers could be found (all P > 0.05). Conclusion The results of this MR analysis did not support that peripheral TNF-α, CRP, IL-1α, IL-1β, IL-2, IL-6, sIL-6Rα, soluble gp130, and IL-8 were causally associated with delirium. More research is needed to explore the role of inflammatory factors in the pathogenesis of delirium.
Collapse
Affiliation(s)
- Miao Yu
- Department of Nursing, Peking University Third Hospital, Beijing, China
| | - Yuxuan Li
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Baohua Li
- Department of Nursing, Peking University Third Hospital, Beijing, China
| | - Qinggang Ge
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| |
Collapse
|
31
|
Langenberg C, Hingorani AD, Whitty CJM. Biological and functional multimorbidity-from mechanisms to management. Nat Med 2023; 29:1649-1657. [PMID: 37464031 DOI: 10.1038/s41591-023-02420-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/23/2023] [Indexed: 07/20/2023]
Abstract
Globally, the number of people with multiple co-occurring diseases will increase substantially over the coming decades, with important consequences for patients, carers, healthcare systems and society. Addressing this challenge requires a shift in the prevailing clinical, educational and scientific thinking and organization-with a strong emphasis on the maintenance of generalist skills to balance the specialization trends of medical education and research. Multimorbidity is not a single entity but differs quantitively and qualitatively across life stages, ethnicities, sexes, socioeconomic groups and geographies. Data-driven science that quantifies the impact of disease co-occurrence-beyond the small number of currently well-studied long-term conditions (such as cardiometabolic diseases)-can help illuminate the pathological diversity of multimorbidity and identify common, mechanistically related, and prognostically relevant clusters. Broader access to data opportunities across modalities and disciplines will catalyze vertical and horizontal integration of multimorbidity research, to enable reconfiguring of medical services, clinical trials, guidelines and research in a way that accounts for the complexity of multimorbidity-and provides efficient, joined-up services for patients.
Collapse
Affiliation(s)
- Claudia Langenberg
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK.
| | - Aroon D Hingorani
- UCL BHF Research Accelerator, University College London, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
- University College London Hospitals NIHR Biomedical Research Centre, London, UK
| | - Christopher J M Whitty
- Department of Health and Social Care, London, UK
- London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
32
|
Weber BN, Giles JT, Liao KP. Shared inflammatory pathways of rheumatoid arthritis and atherosclerotic cardiovascular disease. Nat Rev Rheumatol 2023; 19:417-428. [PMID: 37231248 PMCID: PMC10330911 DOI: 10.1038/s41584-023-00969-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2023] [Indexed: 05/27/2023]
Abstract
The association between chronic inflammation and increased risk of cardiovascular disease in rheumatoid arthritis (RA) is well established. In the general population, inflammation is an established independent risk factor for cardiovascular disease, and much interest is placed on controlling inflammation to reduce cardiovascular events. As inflammation encompasses numerous pathways, the development of targeted therapies in RA provides an opportunity to understand the downstream effect of inhibiting specific pathways on cardiovascular risk. Data from these studies can inform cardiovascular risk management in patients with RA, and in the general population. This Review focuses on pro-inflammatory pathways targeted by existing therapies in RA and with mechanistic data from the general population on cardiovascular risk. Specifically, the discussions include the IL-1, IL-6 and TNF pathways, as well as the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signalling pathway, and the role of these pathways in RA pathogenesis in the joint alongside the development of atherosclerotic cardiovascular disease. Overall, some robust data support inhibition of IL-1 and IL-6 in decreasing the risk of cardiovascular disease, with growing data supporting IL-6 inhibition in both patients with RA and the general population to reduce the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Brittany N Weber
- Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jon T Giles
- Columbia University, Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Katherine P Liao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA.
- Rheumatology Section, VA Boston Medical Center, Boston, MA, USA.
| |
Collapse
|
33
|
Fang X, Mei W, Zeng R, Zou L, Zeng X, Tang S. CIRC_0012535 CONTRIBUTES TO LIPOPOLYSACCHARIDE-INDUCED FETAL LUNG FIBROBLAST APOPTOSIS AND INFLAMMATION TO REGULATE INFANTILE PNEUMONIA DEVELOPMENT BY MODULATING THE MIR-338-3P/IL6R SIGNALING. Shock 2023; 59:820-828. [PMID: 36870073 DOI: 10.1097/shk.0000000000002111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
ABSTRACT Background: Infantile pneumonia is a respiratory infection disease, seriously threatening the life of neonatal patients. Circular RNA (circRNA) dysregulation is reported to be involved in pneumonia pathogenesis. Circ_0012535 was previously displayed to be upregulated in blood samples of patients with community-acquired pneumonia. However, circ_0012535's role in this disorder remains unclear. We thus aim to unveil the functions of circ_0012535 in infantile pneumonia. Methods: Fetal lung fibroblasts (WI38) treated with LPS were used as pneumonia cell models. Expression analysis for circ_0012535, miR-338-3p and IL6R was performed using quantitative real-time polymerase chain reaction. Cell counting kit 88), 5-ethynyl-2'-deoxyuridine, and flow cytometry assays were implemented for cell function detection. The release of inflammatory factors, and superoxide dismutase activity and malonaldehyde content were ascertained using commercial kits. The putative binding between miR-338-3p and circ_0012535 or IL6R was validated by dual-luciferase analysis, RIP analysis, and pull-down analysis. Results: Circ_0012535 was highly expressed in LPS-treated WI38 cells. Knockdown of circ_0012535 recovered LPS-inhibited cell viability and proliferation and attenuated LPS-induced cell apoptosis, cell cycle arrest, inflammation, and oxidative stress. Circ_0012535 bound to miR-338-3p and negatively regulated miR-338-3p expression. Inhibition of miR-338-3p reversed the role of circ_0012535 knockdown, thereby recovering LPS-induced WI38 cell apoptosis and inflammation. MiR-338-3p bound to IL6R 3'UTR, and circ_0012535 shared miR-338-3p binding site with IL6R. IL6R overexpression reversed the role of miR-338-3p, thereby recovering LPS-induced WI38 cell apoptosis and inflammation. Conclusion: Circ_0012535 supported LPS-induced WI38 cell apoptosis and inflammation to promote the progression of infantile pneumonia, and circ_0012535 functioned partly by targeting the miR-338-3p/IL6R signaling.
Collapse
Affiliation(s)
- Xing Fang
- Department of PICU, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | | | | | | | | | | |
Collapse
|
34
|
Shuvalova YA, Kaminnaya V, Kaminnyi AI. Contribution of Interleukin-6 system genes polymorphisms to the development of coronary atherosclerosis. Gene 2023; 861:147253. [PMID: 36754176 DOI: 10.1016/j.gene.2023.147253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/19/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023]
Abstract
Coronary artery disease (CAD) continues to be a main cause of high cardiovascular morbidity and mortality and its prevalence is expected to increase as the population ages, so its prevention is a key public health policy goal. The risk of developing atherosclerosis is related to a complex interaction of genetic, environmental and lifestyle factors. Significant progress has been made in understanding the genetic architecture of this disease in the last decade. In this article, we attempt to map the current knowledge about the genetics of atherosclerosis, in particular the interleukin-6 system and its contribution to the development of coronary atherosclerosis.
Collapse
Affiliation(s)
- Yulia A Shuvalova
- FSBI Academician E I Chazov National Medical Research Centre of Cardiology of the Ministry of Health of the Russian Federation, Department of Atherosclerosis Problems, 3rd Cherepkovskaya str, 15a, Moscow 121552, Russia.
| | - Victoria Kaminnaya
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry, Delegatskaya str, 20/1, Moscow 127473, Russia.
| | - Alexander I Kaminnyi
- FSBI Academician E I Chazov National Medical Research Centre of Cardiology of the Ministry of Health of the Russian Federation, Department of Atherosclerosis Problems, 3rd Cherepkovskaya str, 15a, Moscow 121552, Russia.
| |
Collapse
|
35
|
Ma CS. T-helper-2 cells and atopic disease: lessons learnt from inborn errors of immunity. Curr Opin Immunol 2023; 81:102298. [PMID: 36870225 DOI: 10.1016/j.coi.2023.102298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 03/06/2023]
Abstract
Inborn errors of immunity (IEI) are caused by monogenic variants that affect the host response to bacterial, viral, and fungal pathogens. As such, individuals with IEI often present with severe, recurrent, and life-threatening infections. However, the spectrum of disease due to IEI is very broad and extends to include autoimmunity, malignancy, and atopic diseases such as eczema, atopic dermatitis, and food and environmental allergies. Here, I review IEI that affect cytokine signaling pathways that dysregulate CD4+ T-cell differentiation, resulting in increased T-helper-2 (Th2) cell development, function, and pathogenicity. These are elegant examples of how rare IEI can provide unique insights into more common pathologies such as allergic disease that are impacting the general population at increased frequency.
Collapse
Affiliation(s)
- Cindy S Ma
- Garvan Institute of Medical Research, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia; Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Australia.
| |
Collapse
|
36
|
Hamilton F, Mitchell RE, Constantinescu A, Hughes D, Cunnington A, Ghazal P, Timpson NJ. The effect of interleukin-6 signaling on severe malaria: A Mendelian randomization analysis. Int J Infect Dis 2023; 129:251-259. [PMID: 36801374 PMCID: PMC10728776 DOI: 10.1016/j.ijid.2023.02.008] [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: 11/08/2022] [Revised: 01/19/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
OBJECTIVES Severe malaria remains a deadly disease for many young children in low- and middle-income countries. Levels of interleukin (IL)-6 have been shown to identify cases of severe malaria and associate with severity, but it is unknown if this association is causal. METHODS A single nucleotide polymorphism (SNP; rs2228145) in the IL-6 receptor was chosen as a genetic variant that is known to alter IL-6 signaling. We tested this, then took this forward as an instrument to perform Mendelian randomization (MR) in MalariaGEN, a large cohort study of patients with severe malaria at 11 worldwide sites. RESULTS In MR analyses using rs2228145, we did not identify an effect of decreased IL-6 signaling on severe malaria (odds ratio 1.14, 95% confidence interval 0.56-2.34, P = 0.713). The estimates of the association with any severe malaria subphenotype were similarly null, although with some imprecision. Further analyses using other MR approaches had similar results. CONCLUSION These analyses do not support a causal role for IL-6 signaling in the development of severe malaria. This result suggests IL-6 may not be causal for severe outcomes in malaria, and that therapeutic manipulation of IL-6 is unlikely to be a suitable treatment for severe malaria.
Collapse
Affiliation(s)
- Fergus Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Infection Sciences, North Bristol NHS Trust, Bristol, UK.
| | - Ruth E Mitchell
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | | | - David Hughes
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Aubrey Cunnington
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK; Centre for Paediatrics and Child Health, Imperial College London, London, UK
| | - Peter Ghazal
- System Immunity Research Institute, Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | | |
Collapse
|
37
|
Coskun A, Zarepour A, Zarrabi A. Physiological Rhythms and Biological Variation of Biomolecules: The Road to Personalized Laboratory Medicine. Int J Mol Sci 2023; 24:ijms24076275. [PMID: 37047252 PMCID: PMC10094461 DOI: 10.3390/ijms24076275] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
The concentration of biomolecules in living systems shows numerous systematic and random variations. Systematic variations can be classified based on the frequency of variations as ultradian (<24 h), circadian (approximately 24 h), and infradian (>24 h), which are partly predictable. Random biological variations are known as between-subject biological variations that are the variations among the set points of an analyte from different individuals and within-subject biological variation, which is the variation of the analyte around individuals’ set points. The random biological variation cannot be predicted but can be estimated using appropriate measurement and statistical procedures. Physiological rhythms and random biological variation of the analytes could be considered the essential elements of predictive, preventive, and particularly personalized laboratory medicine. This systematic review aims to summarize research that have been done about the types of physiological rhythms, biological variations, and their effects on laboratory tests. We have searched the PubMed and Web of Science databases for biological variation and physiological rhythm articles in English without time restrictions with the terms “Biological variation, Within-subject biological variation, Between-subject biological variation, Physiological rhythms, Ultradian rhythms, Circadian rhythm, Infradian rhythms”. It was concluded that, for effective management of predicting, preventing, and personalizing medicine, which is based on the safe and valid interpretation of patients’ laboratory test results, both physiological rhythms and biological variation of the measurands should be considered simultaneously.
Collapse
|
38
|
Arends CM, Liman TG, Strzelecka PM, Kufner A, Löwe P, Huo S, Stein CM, Piper SK, Tilgner M, Sperber PS, Dimitriou S, Heuschmann PU, Hablesreiter R, Harms C, Bullinger L, Weber JE, Endres M, Damm F. Associations of clonal hematopoiesis with recurrent vascular events and death in patients with incident ischemic stroke. Blood 2023; 141:787-799. [PMID: 36441964 DOI: 10.1182/blood.2022017661] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Clonal hematopoiesis (CH) is common among older people and is associated with an increased risk of atherosclerosis, inflammation, and shorter overall survival. Age and inflammation are major risk factors for ischemic stroke, yet the association of CH with risk of secondary vascular events and death is unknown. We investigated CH in peripheral blood DNA from 581 patients with first-ever ischemic stroke from the Prospective Cohort With Incident Stroke-Berlin study using error-corrected targeted sequencing. The primary composite end point (CEP) consisted of recurrent stroke, myocardial infarction, and all-cause mortality. A total of 348 somatic mutations with a variant allele frequency ≥1% were identified in 236 of 581 patients (41%). CH was associated with large-artery atherosclerosis stroke (P = .01) and white matter lesion (P < .001). CH-positive patients showed increased levels of proinflammatory cytokines, such as interleukin-6 (IL-6), interferon gamma, high-sensitivity C-reactive protein, and vascular cell adhesion molecule 1. CH-positive patients had a higher risk for the primary CEP (hazard ratio [HR], 1.55; 95% confidence interval [CI], 1.04-2.31; P = .03), which was more pronounced in patients with larger clones. CH clone size remained an independent risk factor (HR, 1.30; 95% CI, 1.04-1.62; P = .022) in multivariable Cox regression. Although our data show that, in particular, larger and TET2- or PPM1D-mutated clones are associated with increased risk of recurrent vascular events and death, this risk is partially mitigated by a common germline variant of the IL-6 receptor (IL-6R p.D358A). The CH mutation profile is accompanied by a proinflammatory profile, opening new avenues for preventive precision medicine approaches to resolve the self-perpetuating cycle of inflammation and clonal expansion.
Collapse
Affiliation(s)
- Christopher M Arends
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas G Liman
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen), Partner Site, Berlin, Germany
- German Center for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauferkrankungen), Partner Site, Berlin, Germany
- Department of Neurology, Evangelical Hospital Oldenburg, Carl von Ossietzky-University, Oldenburg, Germany
| | - Paulina M Strzelecka
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anna Kufner
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Pelle Löwe
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Shufan Huo
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Catarina M Stein
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sophie K Piper
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Medical Informatics, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marlon Tilgner
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pia S Sperber
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Savvina Dimitriou
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Peter U Heuschmann
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University of Würzburg, Würzburg, Germany
- Clinical Trial Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Raphael Hablesreiter
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christoph Harms
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauferkrankungen), Partner Site, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK), Partner Site, Berlin, Germany
- German Cancer Research Center (Deutsches Krebsforschungszentrum), Heidelberg, Germany
| | - Joachim E Weber
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthias Endres
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen), Partner Site, Berlin, Germany
- German Center for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauferkrankungen), Partner Site, Berlin, Germany
| | - Frederik Damm
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK), Partner Site, Berlin, Germany
| |
Collapse
|
39
|
Hamilton F, Schurz H, Yates TA, Gilchrist JJ, Möller M, Naranbhai V, Ghazal P, Timpson NJ, Parks T, Pollara G. Altered IL-6 signalling and risk of tuberculosis disease: a meta-analysis and Mendelian randomisation study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.07.23285472. [PMID: 36798349 PMCID: PMC9934798 DOI: 10.1101/2023.02.07.23285472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
IL-6 responses are ubiquitous in Mycobacterium tuberculosis (Mtb) infections, but their role in determining human tuberculosis (TB) disease risk is unknown. We used single nucleotide polymorphisms (SNPs) in and near the IL-6 receptor (IL6R) gene, focusing on the non-synonymous variant, rs2228145, associated with reduced classical IL-6 signalling, to assess the effect of altered IL-6 activity on TB disease risk. We identified 16 genome wide association studies (GWAS) of TB disease collating 17,982 cases of TB disease and 972,389 controls across 4 continents. Meta-analyses and Mendelian randomisation analyses revealed that reduced classical IL-6 signalling was associated with lower odds of TB disease, a finding replicated using multiple, independent SNP instruments and 2 separate exposure variables. Our findings establish a causal relationship between IL-6 signalling and the outcome of Mtb infection, suggesting IL-6 antagonists do not increase the risk of TB disease and should be investigated as adjuncts in treatment.
Collapse
Affiliation(s)
- Fergus Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Haiko Schurz
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Tom A. Yates
- Division of Infection and Immunity, University College London, London, UK
| | - James J. Gilchrist
- Wellcome Trust Centre for Human Genetics, Oxford, UK
- Department of Paediatrics, University of Oxford, UK
| | - Marlo Möller
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vivek Naranbhai
- Wellcome Trust Centre for Human Genetics, Oxford, UK
- Massachusetts General Hospital, Boston, USA
- Dana-Farber Cancer Institute, Boston, USA
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- Harvard Medical School, Boston, USA
| | | | | | - Tom Parks
- Wellcome Trust Centre for Human Genetics, Oxford, UK
- Department of Infectious Diseases Imperial College London, London, UK
| | - Gabriele Pollara
- Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
40
|
Daghlas I, Gill D. Mendelian randomization as a tool to inform drug development using human genetics. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e16. [PMID: 38550933 PMCID: PMC10953771 DOI: 10.1017/pcm.2023.5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/14/2023] [Accepted: 01/30/2023] [Indexed: 04/11/2024]
Abstract
Drug development is essential to the advancement of human health, however, the process is slow, costly, and at high risk of failure at all stages. A promising strategy for expediting and improving the probability of success in the drug development process is the use of naturally randomized human genetic variation for drug target identification and validation. These data can be harnessed using the Mendelian randomization (MR) analytic paradigm to proxy the lifelong consequences of genetic perturbations of drug targets. In this review, we discuss the myriad applications of the MR paradigm for human drug target identification and validation. We review the methodology and applications of MR, key limitations of MR, and potential future opportunities for research. Throughout the review, we refer to illustrative examples of MR analyses investigating the consequences of genetic inhibition of interleukin 6 signaling which, in some cases, have anticipated results from randomized controlled trials. As human genetic data become more widely available, we predict that MR will serve as a key pillar of support for drug development efforts.
Collapse
Affiliation(s)
- Iyas Daghlas
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Chief Scientific Advisor Office, Research and Early Development, Novo Nordisk, Copenhagen, Denmark
| |
Collapse
|
41
|
Hamilton FW, Thomas M, Arnold D, Palmer T, Moran E, Mentzer AJ, Maskell N, Baillie K, Summers C, Hingorani A, MacGowan A, Khandaker GM, Mitchell R, Davey Smith G, Ghazal P, Timpson NJ. Therapeutic potential of IL6R blockade for the treatment of sepsis and sepsis-related death: A Mendelian randomisation study. PLoS Med 2023; 20:e1004174. [PMID: 36716318 PMCID: PMC9925069 DOI: 10.1371/journal.pmed.1004174] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 02/13/2023] [Accepted: 01/13/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Sepsis is characterised by dysregulated, life-threatening immune responses, which are thought to be driven by cytokines such as interleukin 6 (IL-6). Genetic variants in IL6R known to down-regulate IL-6 signalling are associated with improved Coronavirus Disease 2019 (COVID-19) outcomes, a finding later confirmed in randomised trials of IL-6 receptor antagonists (IL6RAs). We hypothesised that blockade of IL6R could also improve outcomes in sepsis. METHODS AND FINDINGS We performed a Mendelian randomisation (MR) analysis using single nucleotide polymorphisms (SNPs) in and near IL6R to evaluate the likely causal effects of IL6R blockade on sepsis (primary outcome), sepsis severity, other infections, and COVID-19 (secondary outcomes). We weighted SNPs by their effect on CRP and combined results across them in inverse variance weighted meta-analysis, proxying the effect of IL6RA. Our outcomes were measured in UK Biobank, FinnGen, the COVID-19 Host Genetics Initiative (HGI), and the GenOSept and GainS consortium. We performed several sensitivity analyses to test assumptions of our methods, including utilising variants around CRP and gp130 in a similar analysis. In the UK Biobank cohort (N = 486,484, including 11,643 with sepsis), IL6R blockade was associated with a decreased risk of our primary outcome, sepsis (odds ratio (OR) = 0.80; 95% confidence interval (CI) 0.66 to 0.96, per unit of natural log-transformed CRP decrease). The size of this effect increased with severity, with larger effects on 28-day sepsis mortality (OR = 0.74; 95% CI 0.47 to 1.15); critical care admission with sepsis (OR = 0.48, 95% CI 0.30 to 0.78) and critical care death with sepsis (OR = 0.37, 95% CI 0.14 to 0.98). Similar associations were seen with severe respiratory infection: OR for pneumonia in critical care 0.69 (95% CI 0.49 to 0.97) and for sepsis survival in critical care (OR = 0.22; 95% CI 0.04 to 1.31) in the GainS and GenOSept consortium, although this result had a large degree of imprecision. We also confirm the previously reported protective effect of IL6R blockade on severe COVID-19 (OR = 0.69, 95% CI 0.57 to 0.84) in the COVID-19 HGI, which was of similar magnitude to that seen in sepsis. Sensitivity analyses did not alter our primary results. These results are subject to the limitations and assumptions of MR, which in this case reflects interpretation of these SNP effects as causally acting through blockade of IL6R, and reflect lifetime exposure to IL6R blockade, rather than the effect of therapeutic IL6R blockade. CONCLUSIONS IL6R blockade is causally associated with reduced incidence of sepsis. Similar but imprecisely estimated results supported a causal effect also on sepsis related mortality and critical care admission with sepsis. These effects are comparable in size to the effect seen in severe COVID-19, where IL-6 receptor antagonists were shown to improve survival. These data suggest that a randomised trial of IL-6 receptor antagonists in sepsis should be considered.
Collapse
Affiliation(s)
- Fergus W. Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Infection Science, North Bristol NHS Trust, Bristol, United Kingdom
| | - Matt Thomas
- Intensive Care Unit, North Bristol NHS Trust, Bristol, United Kingdom
| | - David Arnold
- Academic Respiratory Unit, University of Bristol, Bristol, United Kingdom
| | - Tom Palmer
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Ed Moran
- Infection Science, North Bristol NHS Trust, Bristol, United Kingdom
| | - Alexander J. Mentzer
- Wellcome Centre For Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Nick Maskell
- Academic Respiratory Unit, University of Bristol, Bristol, United Kingdom
| | - Kenneth Baillie
- Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Charlotte Summers
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Aroon Hingorani
- UCL Institute for Cardiovascular Science, University College London, London, United Kingdom
- UCL BHF Research Accelerator, University College London, London, United Kingdom
- Health Data Research UK, London, United Kingdom
| | | | - Golam M. Khandaker
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Ruth Mitchell
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Peter Ghazal
- Project Sepsis, Cardiff University, Cardiff, United Kingdom
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
42
|
Hizawa N. The understanding of asthma pathogenesis in the era of precision medicine. Allergol Int 2023; 72:3-10. [PMID: 36195530 DOI: 10.1016/j.alit.2022.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/30/2022] [Indexed: 01/25/2023] Open
Abstract
Asthma is a syndrome with extremely diverse clinical phenotypes in which the onset, severity, and response to treatment are defined by the complex interplay of many genetic and environmental factors. Environmental factors epigenetically affect gene expression, and the disease is driven by a multidimensional dynamic network involving RNA and protein molecules derived from gene expression, as well as various metabolic products. In other words, specific pathophysiological mechanisms or endotypes are dynamic networks that arise in response to individual genotypes and the various environmental factors to which individuals have been exposed since before birth, such as diet, infection, air pollution, smoking, antibiotic use, and the bacterial flora of the intestinal tract, skin, and lungs. A key feature of asthma genome scans is their potential to reveal the molecular pathways that lead to pathogenesis. Endotypes that drive the disease have a significant impact on the phenotypes of asthma patients, including their drug responsiveness. Understanding endotypes will lead to not only the implementation of therapies that are tailored to the specific molecular network(s) underlying the patient's condition, but also to the development of therapeutic strategies that target individual endotypes, as well as to precision health, which will enable the prediction of disease onset with high accuracy from an early stage and the implementation of preventive strategies based on endotypes. Understanding of endotypes will pave the way for the practice of precision medicine in asthma care, moving away from 'one-size-fits-all' medicine and population-based prevention approaches that do not take individuals' susceptibility into account.
Collapse
Affiliation(s)
- Nobuyuki Hizawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
43
|
Nash D, Hughes MG, Butcher L, Aicheler R, Smith P, Cullen T, Webb R. IL-6 signaling in acute exercise and chronic training: Potential consequences for health and athletic performance. Scand J Med Sci Sports 2023; 33:4-19. [PMID: 36168944 PMCID: PMC10092579 DOI: 10.1111/sms.14241] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/25/2022] [Accepted: 09/20/2022] [Indexed: 12/13/2022]
Abstract
The cytokine interleukin-6 (IL-6) is involved in a diverse set of physiological processes. Traditionally, IL-6 has been thought of in terms of its inflammatory actions during the acute phase response and in chronic conditions such as rheumatoid arthritis and obesity. However, IL-6 is also an important signaling molecule during exercise, being acutely released from working muscle fibers with increased exercise duration, intensity, and muscle glycogen depletion. In this context, IL-6 enables muscle-organ crosstalk, facilitating a coordinated response to help maintain muscle energy homeostasis, while also having anti-inflammatory actions. The range of actions of IL-6 can be explained by its dichotomous signaling pathways. Classical signaling involves IL-6 binding to a cell-surface receptor (mbIL-6R; present on only a small number of cell types) and is the predominant signaling mechanism during exercise. Trans-signaling involves IL-6 binding to a soluble version of its receptor (sIL-6R), with the resulting complex having a much greater half-life and the ability to signal in all cell types. Trans-signaling drives the inflammatory actions of IL-6 and is the predominant pathway in disease. A single nucleotide polymorphism (rs2228145) on the IL-6R gene can modify the classical/trans-signaling balance through increasing the levels of sIL-6R. This SNP has clinical significance, having been linked to inflammatory conditions such as rheumatoid arthritis and type 1 diabetes, as well as to the severity of symptoms experienced with COVID-19. This review will describe how acute exercise, chronic training and the rs2228145 SNP can modify the IL-6 signaling pathway and the consequent implications for health and athletic performance.
Collapse
Affiliation(s)
- Dan Nash
- School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Michael G Hughes
- School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Lee Butcher
- School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Rebecca Aicheler
- School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Paul Smith
- School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Tom Cullen
- Research Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, UK
| | - Richard Webb
- School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| |
Collapse
|
44
|
Kessler MD, Damask A, O'Keeffe S, Banerjee N, Li D, Watanabe K, Marketta A, Van Meter M, Semrau S, Horowitz J, Tang J, Kosmicki JA, Rajagopal VM, Zou Y, Houvras Y, Ghosh A, Gillies C, Mbatchou J, White RR, Verweij N, Bovijn J, Parikshak NN, LeBlanc MG, Jones M, Glass DJ, Lotta LA, Cantor MN, Atwal GS, Locke AE, Ferreira MAR, Deering R, Paulding C, Shuldiner AR, Thurston G, Ferrando AA, Salerno W, Reid JG, Overton JD, Marchini J, Kang HM, Baras A, Abecasis GR, Jorgenson E. Common and rare variant associations with clonal haematopoiesis phenotypes. Nature 2022; 612:301-309. [PMID: 36450978 PMCID: PMC9713173 DOI: 10.1038/s41586-022-05448-9] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 10/14/2022] [Indexed: 12/02/2022]
Abstract
Clonal haematopoiesis involves the expansion of certain blood cell lineages and has been associated with ageing and adverse health outcomes1-5. Here we use exome sequence data on 628,388 individuals to identify 40,208 carriers of clonal haematopoiesis of indeterminate potential (CHIP). Using genome-wide and exome-wide association analyses, we identify 24 loci (21 of which are novel) where germline genetic variation influences predisposition to CHIP, including missense variants in the lymphocytic antigen coding gene LY75, which are associated with reduced incidence of CHIP. We also identify novel rare variant associations with clonal haematopoiesis and telomere length. Analysis of 5,041 health traits from the UK Biobank (UKB) found relationships between CHIP and severe COVID-19 outcomes, cardiovascular disease, haematologic traits, malignancy, smoking, obesity, infection and all-cause mortality. Longitudinal and Mendelian randomization analyses revealed that CHIP is associated with solid cancers, including non-melanoma skin cancer and lung cancer, and that CHIP linked to DNMT3A is associated with the subsequent development of myeloid but not lymphoid leukaemias. Additionally, contrary to previous findings from the initial 50,000 UKB exomes6, our results in the full sample do not support a role for IL-6 inhibition in reducing the risk of cardiovascular disease among CHIP carriers. Our findings demonstrate that CHIP represents a complex set of heterogeneous phenotypes with shared and unique germline genetic causes and varied clinical implications.
Collapse
Affiliation(s)
| | - Amy Damask
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | - Dadong Li
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | | | | | | | - Jing Tang
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | - Yuxin Zou
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Hyun M Kang
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Aris Baras
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | |
Collapse
|
45
|
Kornej J, Qadan MA, Alotaibi M, Van Wagoner DR, Watrous JD, Trinquart L, Preis SR, Ko D, Jain M, Benjamin EJ, Cheng S, Lin H. The association between eicosanoids and incident atrial fibrillation in the Framingham Heart Study. Sci Rep 2022; 12:20218. [PMID: 36418854 PMCID: PMC9684401 DOI: 10.1038/s41598-022-21786-0] [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: 06/23/2022] [Accepted: 10/04/2022] [Indexed: 11/26/2022] Open
Abstract
Chronic inflammation is a continuous low-grade activation of the systemic immune response. Whereas downstream inflammatory markers are associated with atrial fibrillation (AF), upstream inflammatory effectors including eicosanoids are less studied. To examine the association between eicosanoids and incident AF. We used a liquid chromatography-mass spectrometry for the non-targeted measurement of 161 eicosanoids and eicosanoid-related metabolites in the Framingham Heart Study. The association of each eicosanoid and incident AF was assessed using Cox proportional hazards models and adjusted for AF risk factors, including age, sex, height, weight, systolic/diastolic blood pressure, current smoking, antihypertensive medication, diabetes, history of myocardial infarction and heart failure. False discovery rate (FDR) was used to adjust for multiple testing. Eicosanoids with FDR < 0.05 were considered significant. In total, 2676 AF-free individuals (mean age 66 ± 9 years, 56% females) were followed for mean 10.8 ± 3.4 years; 351 participants developed incident AF. Six eicosanoids were associated with incident AF after adjusting for multiple testing (FDR < 0.05). A joint score was built from the top eicosanoids weighted by their effect sizes, which was associated with incident AF (HR = 2.72, CI = 1.71-4.31, P = 2.1 × 10-5). In conclusion, six eicosanoids were associated with incident AF after adjusting for clinical risk factors for AF.
Collapse
Affiliation(s)
- Jelena Kornej
- National Heart, Lung, and Blood Institute, Boston University's Framingham Heart Study, Framingham, MA, USA. .,Section of Cardiovascular Medicine, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA.
| | - Maha A. Qadan
- grid.239578.20000 0001 0675 4725Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH USA
| | - Mona Alotaibi
- grid.266100.30000 0001 2107 4242Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, CA USA
| | - David R. Van Wagoner
- grid.239578.20000 0001 0675 4725Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH USA
| | - Jeramie D. Watrous
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California, La Jolla, San Diego, CA USA
| | - Ludovic Trinquart
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Sarah R. Preis
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Darae Ko
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.189504.10000 0004 1936 7558Section of Cardiovascular Medicine, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA USA
| | - Mohit Jain
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California, La Jolla, San Diego, CA USA
| | - Emelia J. Benjamin
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.189504.10000 0004 1936 7558Section of Cardiovascular Medicine, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA USA ,grid.189504.10000 0004 1936 7558Department of Epidemiology, Boston University School of Public Health, Boston, MA USA
| | - Susan Cheng
- grid.512369.aDepartment of Cardiology, Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA USA
| | - Honghuang Lin
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.168645.80000 0001 0742 0364Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA USA
| |
Collapse
|
46
|
Castillo-Avila RG, Genis-Mendoza AD, Juárez-Rojop IE, López-Narváez ML, Dionisio-García DM, Nolasco-Rosales GA, Ramos-Méndez MÁ, Hernández-Díaz Y, Tovilla-Zárate CA, González-Castro TB, Nicolini H. High Serum Levels of IL-6 Are Associated with Suicide Attempt but Not with High Lethality Suicide Attempts: A Preliminary Case-Control Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14735. [PMID: 36429454 PMCID: PMC9690459 DOI: 10.3390/ijerph192214735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 06/16/2023]
Abstract
Suicide attempts are an emerging health problem around the world. Increased levels of IL-6 have been associated with suicidal behavior. Therefore, the aims of this study were to evaluate the serum levels of IL-6 in individuals with suicide attempts and a comparison group and to associate the IL-6 levels with the lethality of the suicide attempt. Additionally, we associated the rs2228145 polymorphism of the IL6R gene with suicide attempts or with the IL-6 serum levels. Suicide attempts and their lethality were evaluated using the Columbia Suicide Severity Rating Scale. The serum concentrations of IL-6 were measured by the ELISA technique in individuals with suicide attempts and then compared to a control group. The rs2228145 polymorphism of the IL6R gene was analyzed by real-time polymerase chain reaction. We found elevated serum levels of IL-6 in the suicide attempt group when compared to the control group (F = 10.37, p = 0.002). However, we found no differences of the IL-6 levels between high and low lethality. The IL6R gene polymorphism rs2479409 was not associated with suicide attempts. Our data suggest that IL-6 serum is increased in individuals with suicide attempts.
Collapse
Affiliation(s)
| | - Alma Delia Genis-Mendoza
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Ciudad de Mexico 14610, Mexico
| | - Isela Esther Juárez-Rojop
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa 86100, Mexico
| | - María Lilia López-Narváez
- Hospital Chiapas Nos Une “Dr. Gilberto Gómez Maza”, Secretaría de Salud, Tuxtla Gutierrez 29045, Mexico
| | - Diana María Dionisio-García
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa 86100, Mexico
| | | | - Miguel Ángel Ramos-Méndez
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa 86100, Mexico
| | - Yazmín Hernández-Díaz
- División Académica Multidisciplinaria de Jalpa de Méndez, Universidad Juárez Autónoma de Tabasco, Jalpa de Mendez 86040, Mexico
| | - Carlos Alfonso Tovilla-Zárate
- División Académica Multidisciplinaria de Comalcalco, Universidad Juárez Autónoma de Tabasco, Comalcalco 86040, Mexico
| | - Thelma Beatriz González-Castro
- División Académica Multidisciplinaria de Jalpa de Méndez, Universidad Juárez Autónoma de Tabasco, Jalpa de Mendez 86040, Mexico
| | - Humberto Nicolini
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Ciudad de Mexico 14610, Mexico
| |
Collapse
|
47
|
Ishigaki K, Sakaue S, Terao C, Luo Y, Sonehara K, Yamaguchi K, Amariuta T, Too CL, Laufer VA, Scott IC, Viatte S, Takahashi M, Ohmura K, Murasawa A, Hashimoto M, Ito H, Hammoudeh M, Emadi SA, Masri BK, Halabi H, Badsha H, Uthman IW, Wu X, Lin L, Li T, Plant D, Barton A, Orozco G, Verstappen SMM, Bowes J, MacGregor AJ, Honda S, Koido M, Tomizuka K, Kamatani Y, Tanaka H, Tanaka E, Suzuki A, Maeda Y, Yamamoto K, Miyawaki S, Xie G, Zhang J, Amos CI, Keystone E, Wolbink G, van der Horst-Bruinsma I, Cui J, Liao KP, Carroll RJ, Lee HS, Bang SY, Siminovitch KA, de Vries N, Alfredsson L, Rantapää-Dahlqvist S, Karlson EW, Bae SC, Kimberly RP, Edberg JC, Mariette X, Huizinga T, Dieudé P, Schneider M, Kerick M, Denny JC, Matsuda K, Matsuo K, Mimori T, Matsuda F, Fujio K, Tanaka Y, Kumanogoh A, Traylor M, Lewis CM, Eyre S, Xu H, Saxena R, Arayssi T, Kochi Y, Ikari K, Harigai M, Gregersen PK, Yamamoto K, Louis Bridges S, Padyukov L, Martin J, Klareskog L, Okada Y, Raychaudhuri S. Multi-ancestry genome-wide association analyses identify novel genetic mechanisms in rheumatoid arthritis. Nat Genet 2022; 54:1640-1651. [PMID: 36333501 PMCID: PMC10165422 DOI: 10.1038/s41588-022-01213-w] [Citation(s) in RCA: 150] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
Rheumatoid arthritis (RA) is a highly heritable complex disease with unknown etiology. Multi-ancestry genetic research of RA promises to improve power to detect genetic signals, fine-mapping resolution and performances of polygenic risk scores (PRS). Here, we present a large-scale genome-wide association study (GWAS) of RA, which includes 276,020 samples from five ancestral groups. We conducted a multi-ancestry meta-analysis and identified 124 loci (P < 5 × 10-8), of which 34 are novel. Candidate genes at the novel loci suggest essential roles of the immune system (for example, TNIP2 and TNFRSF11A) and joint tissues (for example, WISP1) in RA etiology. Multi-ancestry fine-mapping identified putatively causal variants with biological insights (for example, LEF1). Moreover, PRS based on multi-ancestry GWAS outperformed PRS based on single-ancestry GWAS and had comparable performance between populations of European and East Asian ancestries. Our study provides several insights into the etiology of RA and improves the genetic predictability of RA.
Collapse
Affiliation(s)
- Kazuyoshi Ishigaki
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Saori Sakaue
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- The Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yang Luo
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Kyuto Sonehara
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Kensuke Yamaguchi
- Department of Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tiffany Amariuta
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Chun Lai Too
- Immunogenetics Unit, Allergy and Immunology Research Center, Institute for Medical Research, National Institutes of Health Complex, Ministry of Health, Kuala Lumpur, Malaysia
- Department of Medicine, Division of Rheumatology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Vincent A Laufer
- Department of Clinical Immunology and Rheumatology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
- Department of Pathology, Michigan Medicine, Ann Arbor, MI, USA
| | - Ian C Scott
- Haywood Academic Rheumatology Centre, Haywood Hospital, Midlands Partnership NHS Foundation Trust, Burslem, UK
- Primary Care Centre Versus Arthritis, School of Medicine, Keele University, Keele, UK
| | - Sebastien Viatte
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester University Foundation Trust, Manchester, UK
| | - Meiko Takahashi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Murasawa
- Department of Rheumatology, Niigata Rheumatic Center, Niigata, Japan
| | - Motomu Hashimoto
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Clinical Immunology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hiromu Ito
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Orthopaedic Surgery, Kurashiki Central Hospital, Kurashiki, Japan
| | - Mohammed Hammoudeh
- Rheumatology Division, Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Samar Al Emadi
- Rheumatology Division, Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Basel K Masri
- Department of Internal Medicine, Jordan Hospital, Amman, Jordan
| | - Hussein Halabi
- Section of Rheumatology, Department of Internal Medicine, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Humeira Badsha
- Dr. Humeira Badsha Medical Center, Emirates Hospital, Dubai, United Arab Emirates
| | - Imad W Uthman
- Department of Rheumatology, American University of Beirut, Beirut, Lebanon
| | - Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzeng Hospital, The Second Military Medical University, Shanghai, China
| | - Li Lin
- Department of Rheumatology and Immunology, Shanghai Changzeng Hospital, The Second Military Medical University, Shanghai, China
| | - Ting Li
- Department of Rheumatology and Immunology, Shanghai Changzeng Hospital, The Second Military Medical University, Shanghai, China
| | - Darren Plant
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester University Foundation Trust, Manchester, UK
| | - Gisela Orozco
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester University Foundation Trust, Manchester, UK
| | - Suzanne M M Verstappen
- NIHR Manchester Biomedical Research Centre, Manchester University Foundation Trust, Manchester, UK
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, UK
| | - John Bowes
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester University Foundation Trust, Manchester, UK
| | | | - Suguru Honda
- Institute of Rheumatology, Tokyo Women's Medical University Hospital, Tokyo, Japan
- Department of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Masaru Koido
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kohei Tomizuka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroaki Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Eiichi Tanaka
- Institute of Rheumatology, Tokyo Women's Medical University Hospital, Tokyo, Japan
- Department of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yuichi Maeda
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoru Miyawaki
- Department of Neurosurgery, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Gang Xie
- Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Jinyi Zhang
- Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Gertjan Wolbink
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center (ARC), Reade, Amsterdam, the Netherlands
| | - Irene van der Horst-Bruinsma
- Department of Rheumatology & Clinical Immunology/ARC, Amsterdam Institute for Infection and Immunity, Amsterdam UMC location Vrije Universiteit, Amsterdam, the Netherlands
| | - Jing Cui
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Katherine P Liao
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA
| | - Robert J Carroll
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hye-Soon Lee
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
- Hanyang University Institute for Rheumatology Research, Seoul, Korea
| | - So-Young Bang
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
- Hanyang University Institute for Rheumatology Research, Seoul, Korea
| | - Katherine A Siminovitch
- Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
- Departments of Medicine and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Niek de Vries
- Department of Rheumatology & Clinical Immunology/ARC, Amsterdam Institute for Infection and Immunity, Amsterdam UMC location AMC/University of Amsterdam, Amsterdam, the Netherlands
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Elizabeth W Karlson
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
- Hanyang University Institute for Rheumatology Research, Seoul, Korea
| | - Robert P Kimberly
- Center for Clinical and Translational Science, Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeffrey C Edberg
- Center for Clinical and Translational Science, Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xavier Mariette
- Department of Rheumatology, Université Paris-Saclay, Assistance Pubique - Hôpitaux de Paris, Hôpital Bicêtre, INSERM UMR1184, Le Kremlin Bicêtre, France
| | - Tom Huizinga
- Leiden University Medical Center, Leiden, the Netherlands
| | - Philippe Dieudé
- University of Paris Cité, Inserm, PHERE, F-75018, Paris, France
- Department of Rheumatology, Hôpital Bichat, APHP, Paris, France
| | - Matthias Schneider
- Department of Rheumatology & Hiller Research Unit Rheumatology, UKD, Heinrich-Heine University, Düsseldorf, Germany
| | - Martin Kerick
- Institute of Parasitology and Biomedicine Lopez-Neyra, CSIC, Granada, Spain
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
- All of Us Research Program, Office of the Director, National Institutes of Health, Bethesda, MD, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Koichi Matsuda
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsuneyo Mimori
- Department of Rheumatology and Clinical immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Atsushi Kumanogoh
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
| | - Matthew Traylor
- Department of Medical & Molecular Genetics, King's College London, London, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
- Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Cathryn M Lewis
- Department of Medical & Molecular Genetics, King's College London, London, UK
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
| | - Stephen Eyre
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester University Foundation Trust, Manchester, UK
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzeng Hospital, The Second Military Medical University, Shanghai, China
- School of Clinical Medicine Tsinghua University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
| | - Richa Saxena
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Thurayya Arayssi
- Department of Internal Medicine, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | - Yuta Kochi
- Department of Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Katsunori Ikari
- Institute of Rheumatology, Tokyo Women's Medical University Hospital, Tokyo, Japan
- Department of Orthopedic Surgery, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
- Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women's Medical University, Tokyo, Japan
| | - Masayoshi Harigai
- Institute of Rheumatology, Tokyo Women's Medical University Hospital, Tokyo, Japan
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Peter K Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - S Louis Bridges
- Department of Medicine, Hospital for Special Surgery, New York, NY, USA
- Division of Rheumatology, Weill Cornell Medicine, New York, NY, USA
| | - Leonid Padyukov
- Department of Medicine, Division of Rheumatology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Javier Martin
- Institute of Parasitology and Biomedicine Lopez-Neyra, CSIC, Granada, Spain
| | - Lars Klareskog
- Department of Medicine, Division of Rheumatology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan.
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan.
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan.
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
48
|
Chen L, Peters JE, Prins B, Persyn E, Traylor M, Surendran P, Karthikeyan S, Yonova-Doing E, Di Angelantonio E, Roberts DJ, Watkins NA, Ouwehand WH, Danesh J, Lewis CM, Bronson PG, Markus HS, Burgess S, Butterworth AS, Howson JMM. Systematic Mendelian randomization using the human plasma proteome to discover potential therapeutic targets for stroke. Nat Commun 2022; 13:6143. [PMID: 36253349 PMCID: PMC9576777 DOI: 10.1038/s41467-022-33675-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 09/28/2022] [Indexed: 02/02/2023] Open
Abstract
Stroke is the second leading cause of death with substantial unmet therapeutic needs. To identify potential stroke therapeutic targets, we estimate the causal effects of 308 plasma proteins on stroke outcomes in a two-sample Mendelian randomization framework and assess mediation effects by stroke risk factors. We find associations between genetically predicted plasma levels of six proteins and stroke (P ≤ 1.62 × 10-4). The genetic associations with stroke colocalize (Posterior Probability >0.7) with the genetic associations of four proteins (TFPI, TMPRSS5, CD6, CD40). Mendelian randomization supports atrial fibrillation, body mass index, smoking, blood pressure, white matter hyperintensities and type 2 diabetes as stroke risk factors (P ≤ 0.0071). Body mass index, white matter hyperintensity and atrial fibrillation appear to mediate the TFPI, IL6RA, TMPRSS5 associations with stroke. Furthermore, thirty-six proteins are associated with one or more of these risk factors using Mendelian randomization. Our results highlight causal pathways and potential therapeutic targets for stroke.
Collapse
Affiliation(s)
- Lingyan Chen
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - James E Peters
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, UK
| | - Bram Prins
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Elodie Persyn
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Matthew Traylor
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
- Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, CB1 8RN, Cambridge, UK
| | - Savita Karthikeyan
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Ekaterina Yonova-Doing
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Health Data Science Research Centre, Human Technopole, Milan, Italy
| | - David J Roberts
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant-Oxford Centre, Level 2, John Radcliffe Hospital, Oxford, UK
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Nicholas A Watkins
- NHS Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge, UK
| | - Willem H Ouwehand
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Wellcome Sanger Institute, Hinxton, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Cathryn M Lewis
- Department of Medical and Molecular Genetics, King's College London, London, UK
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
| | | | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Stephen Burgess
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Joanna M M Howson
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK.
| |
Collapse
|
49
|
Sainz L, Riera P, Moya P, Bernal S, Casademont J, Díaz-Torné C, Millán AM, Park HS, Lasa A, Corominas H. Role of IL6R Genetic Variants in Predicting Response to Tocilizumab in Patients with Rheumatoid Arthritis. Pharmaceutics 2022; 14:pharmaceutics14091942. [PMID: 36145690 PMCID: PMC9501307 DOI: 10.3390/pharmaceutics14091942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/21/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis (RA) is a prevalent autoimmune disease characterized by chronic arthritis that may lead to irreversible joint damage and significant disability. Patients with RA are commonly treated with Tocilizumab (TCZ), an IL-6 receptor (IL-6R) antagonist, but many patients refractorily respond to this therapy. Identifying genetic biomarkers as predictors of TCZ response could be a key to providing a personalized medicine strategy. We aimed to evaluate whether functional single nucleotide polymorphisms (SNPs) in the IL6R gene could predict TCZ response in patients with RA. We retrospectively included 88 RA patients treated with TCZ. Six SNPs previously described in the IL6R gene (rs12083537, rs11265618, rs4329505, rs2228145, rs4537545, and rs4845625) were genotyped in DNA samples from these patients. Using parametric tests, we evaluated the association between these polymorphisms and clinicopathological features. Responses to treatments were assessed at six months using three variables: a quantitative improvement in Disease activity score including 28 joints (DAS28), a satisfactory European League Against Rheumatism (EULAR) response, and low disease activity (LDA) achievement. The three response variables studied were associated with genetic variant rs4845625, and no association was found with the other five SNPs. Our findings support the potential clinical value of SNPs in the IL6R gene as predictive biomarkers for TCZ response.
Collapse
Affiliation(s)
- Luis Sainz
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Pau Riera
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (P.R.); (H.C.)
| | - Patricia Moya
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Sara Bernal
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Genetics Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Jordi Casademont
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- Internal Medicine Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Cesar Díaz-Torné
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Ana Milena Millán
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Hye Sang Park
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Adriana Lasa
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Genetics Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Héctor Corominas
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- Correspondence: (P.R.); (H.C.)
| |
Collapse
|
50
|
Forcina L, Franceschi C, Musarò A. The hormetic and hermetic role of IL-6. Ageing Res Rev 2022; 80:101697. [PMID: 35850167 DOI: 10.1016/j.arr.2022.101697] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/24/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
Abstract
Interleukin-6 is a pleiotropic cytokine regulating different tissues and organs in diverse and sometimes discrepant ways. The dual and sometime hermetic nature of IL-6 action has been highlighted in several contexts and can be explained by the concept of hormesis, in which beneficial or toxic effects can be induced by the same molecule depending on the intensity, persistence, and nature of the stimulation. According with hormesis, a low and/or controlled IL-6 release is associated with anti-inflammatory, antioxidant, and pro-myogenic actions, whereas increased systemic levels of IL-6 can induce pro-inflammatory, pro-oxidant and pro-fibrotic responses. However, many aspects regarding the multifaceted action of IL-6 and the complex nature of its signal transduction remains to be fully elucidated. In this review we collect mechanistic insight into the molecular networks contributing to normal or pathologic changes during advancing age and in chronic diseases. We point out the involvement of IL-6 deregulation in aging-related diseases, dissecting the hormetic action of this key mediator in different tissues, with a special focus on skeletal muscle. Since IL-6 can act as an enhancer of detrimental factor associated with both aging and pathologic conditions, such as chronic inflammation and oxidative stress, this cytokine could represent a "Gerokine", a determinant of the switch from physiologic aging to age-related diseases.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, Rome 00161, Italy.
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Scuola Superiore di Studi Avanzati Sapienza (SSAS), Via A. Scarpa, 14, Rome 00161, Italy.
| |
Collapse
|