1
|
Yang B, Chu L, Feng F, Lu S, Xue C. Association of tyrosine kinase 2 polymorphisms with susceptibility to microscopic polyangiitis in a Guangxi population. PeerJ 2024; 12:e18735. [PMID: 39726748 PMCID: PMC11670758 DOI: 10.7717/peerj.18735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Background Heredity and epigenetics affect the pathogenesis of microscopic polyangiitis (MPA). Tyrosine kinase 2 (TYK2) polymorphisms (rs2304256C > A, rs280519A > G, and rs12720270G > A) may be potential protective factors against anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). Current research suggests that TYK2 is associated with various autoimmune diseases; however, no study has examined the relationship between TYK2 polymorphisms and AAV. This study assessed the effect of TYK2 polymorphisms on susceptibility to MPA. Methods Overall, 562 Chinese participants (265 patients with MPA and 297 healthy volunteers) were recruited. Polymerase chain reactions combined with high-throughput sequencing were used to analyze polymorphic loci, while logistic regression analysis was used to assess the relationship between polymorphism of the TYK2 gene and MPA susceptibility. Results In males, individuals with the CA genotype (rs2304256) in the overdominant model showed a significantly reduced risk of MPA (odds ratio (OR) = 0.52; 95% confidence interval (CI) [0.29-0.93]; p = 0.025). Regarding rs280519, male carriers of the AG genotype had a significantly lower risk of developing MPA in both the codominant (OR = 0.51; 95% CI [0.28-0.93]; p = 0.039) and overdominant (OR = 0.48; 95% CI [0.27-0.86]; p = 0.013) models. The GA genotype of rs12720270 was associated with low susceptibility to MPA in males (OR = 0.52; 95% CI [0.29-0.93]; p = 0.027). Conclusions This study indicates that mutations in the TYK2 gene (rs2304256, rs280519, and rs12720270) may be associated with a reduced risk of MPA in the male Chinese population in Guangxi. The A allele of single nucleotide polymorphism (SNP) rs2304256 may be a protective factor against MPA, while the G alleles of SNPs rs280519 and rs12720270 are protective factors against MPA.
Collapse
Affiliation(s)
- Binglan Yang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Liepeng Chu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fei Feng
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shurong Lu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chao Xue
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
2
|
Miranda S, Lassnig C, Schmidhofer K, Kjartansdottir H, Vogl C, Tangermann S, Tsymala I, Babl V, Müller M, Kuchler K, Strobl B. Lack of TYK2 signaling enhances host resistance to Candida albicans skin infection. Nat Commun 2024; 15:10493. [PMID: 39622833 PMCID: PMC11612186 DOI: 10.1038/s41467-024-54888-6] [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: 01/29/2024] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
Candida albicans is the most common human fungal pathogen, causing diseases ranging from local to life-threating systemic infections. Tyrosine kinase 2 (TYK2), a crucial mediator in several cytokine signaling pathways, has been associated with protective functions in various microbial infections. However, its specific contribution in the immune response to fungal infections has remained elusive. In this study, we show that mice lacking TYK2 or its enzymatic activity exhibit enhanced resistance to C. albicans skin infections, limiting fungal spread and accelerating wound healing. Impaired TYK2-signaling prompted the formation of a distinctive layer of necrotic neutrophils around the fungal pathogens. Transcriptomic analysis revealed TYK2's pivotal role in regulating interferon-inducible genes in neutrophils, thereby impacting their antifungal capacity during infection. Furthermore, we show that TYK2-dependent interferon-gamma (IFNγ) production contributes to fungal dissemination from the skin to the kidneys. Our study uncovers a hitherto unrecognized detrimental role of TYK2 in cutaneous C. albicans infections.
Collapse
Affiliation(s)
- Sara Miranda
- Centre of Biological Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Caroline Lassnig
- Centre of Biological Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
- Vetbiomodels, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Kristina Schmidhofer
- Centre of Biological Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hrönn Kjartansdottir
- Centre of Biological Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Claus Vogl
- Centre of Biological Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Simone Tangermann
- Centre of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Irina Tsymala
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Center for Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Verena Babl
- Centre of Biological Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Centre of Biological Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
- Vetbiomodels, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Karl Kuchler
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Center for Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Birgit Strobl
- Centre of Biological Sciences, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Sestan M, Arsov T, Kifer N, Frkovic M, Grguric D, Ellyard J, Cook M, Vinuesa CG, Jelusic M. Whole exome sequencing in patients with childhood-onset systemic lupus erythematosus: Results from a Croatian national study. Scand J Immunol 2024; 100:e13411. [PMID: 39380326 DOI: 10.1111/sji.13411] [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: 02/04/2024] [Revised: 09/15/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024]
Abstract
The purpose of this study was to identify new and low-frequency gene variants using whole exome sequencing (WES) in patients with childhood-onset systemic lupus erythematosus (cSLE), that may be involved in the pathogenesis of SLE. We performed WES on selected 17 trios (in some cases including other informative family members) in which the proband presented with severe, atypical clinical features, resistance to conventional therapy, a family pattern of occurrence and/or syndromic characteristics. After performing WES and analysis of gene variants, 17 novel and/or low-frequency variants were identified in 7 patients. One variant was classified as pathogenic (KMT2D, NM_003482.3:c.8626delC, predicted to truncate the protein p.(Gln2876Serfs*34)) and two as likely pathogenic according to the American College of Medical Genetics and Genomics classification guidelines (ADAR, NM_001111.3:c.2815A>G, predicted to encode p.(Ile939Val); BLK, NM_001715.2:c.211G>A, predicted to encode p.(Ala71Thr)). The other variants remain of uncertain significance at this point of time. WES is an important diagnostic and research instrument, producing a growing list of likely genes and gene variants that may be of relevance in the pathogenesis of cSLE and potentially point to novel therapeutic targets.
Collapse
Affiliation(s)
- Mario Sestan
- Department of Paediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Todor Arsov
- Faculty of Medical Sciences, University Goce Delchev, Shtip, North Macedonia
- The Francis Crick Institute, London, UK
| | - Nastasia Kifer
- Department of Paediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Marijan Frkovic
- Department of Paediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Danica Grguric
- Department of Paediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Julia Ellyard
- Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Matthew Cook
- Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
- University of Cambridge, Cambridge, UK
| | - Carola G Vinuesa
- The Francis Crick Institute, London, UK
- Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Marija Jelusic
- Department of Paediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| |
Collapse
|
4
|
Ucpinar S, Kwan JK, Hoffman JD, Tilley MK, Douglas JA, Rubio RG, Lu R, Nunn PA, Langrish CL. Safety, tolerability, pharmacokinetics, and pharmacodynamics of the oral allosteric TYK2 inhibitor ESK-001 using a randomized, double-blind, placebo-controlled study design. Clin Transl Sci 2024; 17:e70094. [PMID: 39604226 PMCID: PMC11602527 DOI: 10.1111/cts.70094] [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: 03/11/2024] [Revised: 10/25/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
ESK-001 is a highly selective allosteric inhibitor of tyrosine kinase 2 (TYK2), which plays an essential role in mediating cytokine signaling in multiple immune-mediated diseases. In 2 phase I studies, a first-in-human single ascending dose (SAD) and multiple ascending dose (MAD) study and a multiple-dose (MD) study, we evaluated the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of orally administered ESK-001 in healthy participants using a randomized, double-blind, placebo-controlled study design. ESK-001 was rapidly absorbed with systemic exposures generally increasing dose-proportionally across all cohorts. The mean terminal half-life ranged from 8 to 13 h with no to minimal accumulation of ESK-001 following q.d. doses and ~2-fold accumulation following Q12 doses. Less than 1% of unchanged ESK-001 was eliminated in urine. ESK-001 inhibited the downstream TYK2 pathway as shown by inhibition of pSTAT1 expression. Transcriptomic analysis of unstimulated whole blood samples confirmed dose-dependent inhibition of Type I IFN-induced genes and SIGLEC1, a novel TYK2-responsive biomarker. By correlating PK exposure data with PD readouts, a strong PK/PD relationship was demonstrated. There were no deaths, serious treatment-emergent adverse events (TEAEs), nor severe TEAEs, and most TEAEs were mild in severity. In conclusion, ESK-001 was generally safe and well-tolerated in healthy participants, showed linear dose-dependent PK characteristics, and maximally inhibited TYK2-dependent pathways with a predictable concentration-dependent PK/PD relationship. These findings were used to select the dose range of ESK-001 for the STRIDE phase II trial in plaque psoriasis and to support further clinical development of ESK-001 in other TYK2-mediated diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ruixiao Lu
- Alumis IncSouth San FranciscoCaliforniaUSA
| | | | | |
Collapse
|
5
|
Mammoliti O, Menet C, Cottereaux C, Blanc J, De Blieck A, Coti G, Geney R, Oste L, Ostyn K, Palisse A, Quinton E, Schmitt B, Borgonovi M, Parent I, Jagerschmidt C, De Vos S, Vayssiere B, López-Ramos M, Shoji K, Brys R, Amantini D, Galien R, Joannesse C. Design of a potent and selective dual JAK1/TYK2 inhibitor. Bioorg Med Chem 2024; 114:117932. [PMID: 39447537 DOI: 10.1016/j.bmc.2024.117932] [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: 05/07/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024]
Abstract
Janus kinase (JAK) inhibitors have gathered interest as treatments for several inflammatory and autoimmune diseases. The four first marketed inhibitors target JAK1, with varying selectivity towards other JAK family members, but none inhibit tyrosine kinase-2 (TYK2) at clinically relevant doses. TYK2 is required for the signaling of the interleukin (IL)-12 and IL-23 cytokines, which are key to the polarization of TH1 and TH17 cells, respectively; two cell subtypes that play major roles in inflammatory diseases. Herein, we report our effort towards the optimization of a potent and selective dual JAK1/TYK2 inhibitor series starting from a HTS hit. Structural information revealed vectors required to improve both JAK1 and TYK2 potency as well as selectivity towards JAK2. The potent inhibition of both JAK1 (3.5 nM) and TYK2 (5.7 nM) in biochemical assays by our optimized lead compound, as well as its notable selectivity against JAK2, were confirmed in cellular and whole blood assays. Inhibition of TYK2 by the lead compound was demonstrated by dose-dependent efficacy in an IL-23-induced psoriasis-like inflammation mouse model.
Collapse
Affiliation(s)
- Oscar Mammoliti
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Christel Menet
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Céline Cottereaux
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Javier Blanc
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Ann De Blieck
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Ghjuvanni Coti
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Raphaël Geney
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Line Oste
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Koen Ostyn
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Adeline Palisse
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Evelyne Quinton
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Benoit Schmitt
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Monica Borgonovi
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Isabelle Parent
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - Steve De Vos
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | | | | | - Kenji Shoji
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Reginald Brys
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - David Amantini
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - René Galien
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | |
Collapse
|
6
|
Guo X, Tao MJ, Ji X, Han M, Shen Y, Hong C, Guo H, Shi W, Yuan H. Validation of TYK2 and exploration of PRSS36 as drug targets for psoriasis using Mendelian randomization. Sci Rep 2024; 14:23902. [PMID: 39397091 PMCID: PMC11471773 DOI: 10.1038/s41598-024-74148-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
Psoriasis is a chronic inflammatory skin disorder with multiple causes, including genetic and environmental factors. Despite advances in treatment, there remains a need to identify novel therapeutic targets. A Mendelian randomization (MR) analysis was conducted to identify therapeutic targets for psoriasis. Data on cis-expression quantitative trait loci were obtained from the eQTLGen Consortium (n = 31,684). Summary statistics for psoriasis (outcome) were sourced from the GWAS Catalog with a sample size of 484,598, including 5,427 cases and 479,171 controls. Colocalization analysis was used to assess whether psoriasis risk and gene expression were driven by shared single nucleotide polymorphisms. Drug prediction and molecular docking were utilized to validate the pharmacological value of the drug targets. The MR analysis found that 81 drug targets were significantly associated, and two (TYK2 and PRSS36) were supported by colocalization analysis (PP.H4 > 0.80). Phenome-wide association studies did not show any associations with other traits at the gene level. Biologically, these genes were closely related to immune function. Molecular docking revealed strong binding with drugs and proteins, as supported by available structural data. This study validated TYK2 as a drug target for psoriasis, in line with its existing clinical use, including the development of decucravacitinib. PRSS36 is a potential novel target requiring further investigation.
Collapse
Affiliation(s)
- Xin Guo
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu, Anhui, China
| | - Meng-Jun Tao
- Department of Health Management Center, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - XinCan Ji
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu, Anhui, China
| | - MengQi Han
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu, Anhui, China
| | - Yue Shen
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu, Anhui, China
| | - Cheng Hong
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu, Anhui, China
| | - HaoYang Guo
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu, Anhui, China
| | - Wei Shi
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu, Anhui, China.
| | - Hui Yuan
- School of Public Health, Wannan Medical College, No. 22, Wenchang West Road, Yijiang District, Wuhu, Anhui, China.
| |
Collapse
|
7
|
Saadat I, Saadat M. Autoimmune diseases share a common genetic architecture involving the JAK-STAT pathway. EXCLI JOURNAL 2024; 23:1027-1029. [PMID: 39421031 PMCID: PMC11483264 DOI: 10.17179/excli2024-7684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 08/20/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Iraj Saadat
- Department of Biology, School of Science, Shiraz University, Shiraz 71467-13565, Iran
| | - Mostafa Saadat
- Department of Biology, School of Science, Shiraz University, Shiraz 71467-13565, Iran
| |
Collapse
|
8
|
Lv Y, Qi J, Babon JJ, Cao L, Fan G, Lang J, Zhang J, Mi P, Kobe B, Wang F. The JAK-STAT pathway: from structural biology to cytokine engineering. Signal Transduct Target Ther 2024; 9:221. [PMID: 39169031 PMCID: PMC11339341 DOI: 10.1038/s41392-024-01934-w] [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: 04/08/2024] [Revised: 06/12/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway serves as a paradigm for signal transduction from the extracellular environment to the nucleus. It plays a pivotal role in physiological functions, such as hematopoiesis, immune balance, tissue homeostasis, and surveillance against tumors. Dysregulation of this pathway may lead to various disease conditions such as immune deficiencies, autoimmune diseases, hematologic disorders, and cancer. Due to its critical role in maintaining human health and involvement in disease, extensive studies have been conducted on this pathway, ranging from basic research to medical applications. Advances in the structural biology of this pathway have enabled us to gain insights into how the signaling cascade operates at the molecular level, laying the groundwork for therapeutic development targeting this pathway. Various strategies have been developed to restore its normal function, with promising therapeutic potential. Enhanced comprehension of these molecular mechanisms, combined with advances in protein engineering methodologies, has allowed us to engineer cytokines with tailored properties for targeted therapeutic applications, thereby enhancing their efficiency and safety. In this review, we outline the structural basis that governs key nodes in this pathway, offering a comprehensive overview of the signal transduction process. Furthermore, we explore recent advances in cytokine engineering for therapeutic development in this pathway.
Collapse
Affiliation(s)
- You Lv
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100080, China
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Guohuang Fan
- Immunophage Biotech Co., Ltd, No. 10 Lv Zhou Huan Road, Shanghai, 201112, China
| | - Jiajia Lang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jin Zhang
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Pengbing Mi
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland, 4072, Australia.
| | - Faming Wang
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
9
|
Mammoliti O, Martina S, Claes P, Coti G, Blanque R, Jagerschmidt C, Shoji K, Borgonovi M, De Vos S, Marsais F, Oste L, Quinton E, López-Ramos M, Amantini D, Brys R, Jimenez JM, Galien R, van der Plas S. Discovery of GLPG3667, a Selective ATP Competitive Tyrosine Kinase 2 Inhibitor for the Treatment of Autoimmune Diseases. J Med Chem 2024; 67:8545-8568. [PMID: 38805213 PMCID: PMC11181332 DOI: 10.1021/acs.jmedchem.4c00769] [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: 04/02/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024]
Abstract
Tyrosine kinase 2 (TYK2) mediates cytokine signaling through type 1 interferon, interleukin (IL)-12/IL-23, and the IL-10 family. There appears to be an association between TYK2 genetic variants and inflammatory conditions, and clinical evidence suggests that selective inhibition of TYK2 could produce a unique therapeutic profile. Here, we describe the discovery of compound 9 (GLPG3667), a reversible and selective TYK2 adenosine triphosphate competitive inhibitor in development for the treatment of inflammatory and autoimmune diseases. The preclinical pharmacokinetic profile was favorable, and TYK2 selectivity was confirmed in peripheral blood mononuclear cells and whole blood assays. Dermal ear inflammation was reduced in an IL-23-induced in vivo mouse model of psoriasis. GLPG3667 also completed a phase 1b study (NCT04594928) in patients with moderate-to-severe psoriasis where clinical effect was shown within the 4 weeks of treatment and it is now in phase 2 trials for the treatment of dermatomyositis (NCT05695950) and systemic lupus erythematosus (NCT05856448).
Collapse
Affiliation(s)
- Oscar Mammoliti
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | | | - Pieter Claes
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Ghjuvanni Coti
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Roland Blanque
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - Kenji Shoji
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Monica Borgonovi
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Steve De Vos
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Florence Marsais
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Line Oste
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Evelyne Quinton
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - David Amantini
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Reginald Brys
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | | | - René Galien
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | |
Collapse
|
10
|
König LE, Rodriguez S, Hug C, Daneshvari S, Chung A, Bradshaw GA, Sahin A, Zhou G, Eisert RJ, Piccioni F, Das S, Kalocsay M, Sokolov A, Sorger P, Root DE, Albers MW. TYK2 as a novel therapeutic target in Alzheimer's Disease with TDP-43 inclusions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.595773. [PMID: 38895380 PMCID: PMC11185596 DOI: 10.1101/2024.06.04.595773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Neuroinflammation is a pathological feature of many neurodegenerative diseases, including Alzheimer's disease (AD)1,2 and amyotrophic lateral sclerosis (ALS)3, raising the possibility of common therapeutic targets. We previously established that cytoplasmic double-stranded RNA (cdsRNA) is spatially coincident with cytoplasmic pTDP-43 inclusions in neurons of patients with C9ORF72-mediated ALS4. CdsRNA triggers a type-I interferon (IFN-I)-based innate immune response in human neural cells, resulting in their death4. Here, we report that cdsRNA is also spatially coincident with pTDP-43 cytoplasmic inclusions in brain cells of patients with AD pathology and that type-I interferon response genes are significantly upregulated in brain regions affected by AD. We updated our machine-learning pipeline DRIAD-SP (Drug Repurposing In Alzheimer's Disease with Systems Pharmacology) to incorporate cryptic exon (CE) detection as a proxy of pTDP-43 inclusions and demonstrated that the FDA-approved JAK inhibitors baricitinib and ruxolitinib that block interferon signaling show a protective signal only in cortical brain regions expressing multiple CEs. Furthermore, the JAK family member TYK2 was a top hit in a CRISPR screen of cdsRNA-mediated death in differentiated human neural cells. The selective TYK2 inhibitor deucravacitinib, an FDA-approved drug for psoriasis, rescued toxicity elicited by cdsRNA. Finally, we identified CCL2, CXCL10, and IL-6 as candidate predictive biomarkers for cdsRNA-related neurodegenerative diseases. Together, we find parallel neuroinflammatory mechanisms between TDP-43 associated-AD and ALS and nominate TYK2 as a possible disease-modifying target of these incurable neurodegenerative diseases.
Collapse
Affiliation(s)
- Laura E. König
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital,
114 16 Street, Charlestown, MA 02129, USA
| | - Steve Rodriguez
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital,
114 16 Street, Charlestown, MA 02129, USA
| | - Clemens Hug
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
| | - Shayda Daneshvari
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital,
114 16 Street, Charlestown, MA 02129, USA
| | - Alexander Chung
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital,
114 16 Street, Charlestown, MA 02129, USA
| | - Gary A. Bradshaw
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
| | - Asli Sahin
- Department of Neurology, Massachusetts General Hospital,
114 16 Street, Charlestown, MA 02129, USA
| | - George Zhou
- Department of Neurology, Massachusetts General Hospital,
114 16 Street, Charlestown, MA 02129, USA
| | - Robyn J. Eisert
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
| | - Federica Piccioni
- Broad Institute of MIT and Harvard, 75 Ames Street,
Cambridge, MA 02142, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital,
114 16 Street, Charlestown, MA 02129, USA
| | - Marian Kalocsay
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
| | - Artem Sokolov
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
| | - Peter Sorger
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
| | - David E. Root
- Broad Institute of MIT and Harvard, 75 Ames Street,
Cambridge, MA 02142, USA
| | - Mark W. Albers
- Laboratory of Systems Pharmacology, Harvard Program in
Therapeutic Science, Harvard Medical School, Armenise 132, 200 Longwood Avenue,
Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital,
114 16 Street, Charlestown, MA 02129, USA
| |
Collapse
|
11
|
Lv Y, Mi P, Babon JJ, Fan G, Qi J, Cao L, Lang J, Zhang J, Wang F, Kobe B. Small molecule drug discovery targeting the JAK-STAT pathway. Pharmacol Res 2024; 204:107217. [PMID: 38777110 DOI: 10.1016/j.phrs.2024.107217] [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: 03/22/2024] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway functions as a central hub for transmitting signals from more than 50 cytokines, playing a pivotal role in maintaining hematopoiesis, immune balance, and tissue homeostasis. Dysregulation of this pathway has been implicated in various diseases, including immunodeficiency, autoimmune conditions, hematological disorders, and certain cancers. Proteins within this pathway have emerged as effective therapeutic targets for managing these conditions, with various approaches developed to modulate key nodes in the signaling process, spanning from receptor engagement to transcription factor activation. Following the success of JAK inhibitors such as tofacitinib for RA treatment and ruxolitinib for managing primary myelofibrosis, the pharmaceutical industry has obtained approvals for over 10 small molecule drugs targeting the JAK-STAT pathway and many more are at various stages of clinical trials. In this review, we consolidate key strategies employed in drug discovery efforts targeting this pathway, with the aim of contributing to the collective understanding of small molecule interventions in the context of JAK-STAT signaling. We aspire that our endeavors will contribute to advancing the development of innovative and efficacious treatments for a range of diseases linked to this pathway dysregulation.
Collapse
Affiliation(s)
- You Lv
- Center for Molecular Biosciences and Non-Communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi 710054, China; Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi 710026, China
| | - Pengbing Mi
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Guohuang Fan
- Immunophage Biotech Co., Ltd, No. 10 Lv Zhou Huan Road, Shanghai 201112, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100080, China
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Jiajia Lang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jin Zhang
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi 710026, China
| | - Faming Wang
- Center for Molecular Biosciences and Non-Communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi 710054, China.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
12
|
Syed F, Ballew O, Lee CC, Rana J, Krishnan P, Castela A, Weaver SA, Chalasani NS, Thomaidou SF, Demine S, Chang G, Coomans de Brachène A, Alvelos MI, Marselli L, Orr K, Felton JL, Liu J, Marchetti P, Zaldumbide A, Scheuner D, Eizirik DL, Evans-Molina C. Pharmacological inhibition of tyrosine protein-kinase 2 reduces islet inflammation and delays type 1 diabetes onset in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585925. [PMID: 38766166 PMCID: PMC11100605 DOI: 10.1101/2024.03.20.585925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Tyrosine protein-kinase 2 (TYK2), a member of the Janus kinase family, mediates inflammatory signaling through multiple cytokines, including interferon-α (IFNα), interleukin (IL)-12, and IL-23. Missense mutations in TYK2 are associated with protection against type 1 diabetes (T1D), and inhibition of TYK2 shows promise in the management of other autoimmune conditions. Here, we evaluated the effects of specific TYK2 inhibitors (TYK2is) in pre-clinical models of T1D. First, human β cells, cadaveric donor islets, and iPSC-derived islets were treated in vitro with IFNα in combination with a small molecule TYK2i (BMS-986165 or a related molecule BMS-986202). TYK2 inhibition prevented IFNα-induced β cell HLA class I up-regulation, endoplasmic reticulum stress, and chemokine production. In co-culture studies, pre-treatment of β cells with a TYK2i prevented IFNα-induced activation of T cells targeting an epitope of insulin. In vivo administration of BMS-986202 in two mouse models of T1D (RIP-LCMV-GP mice and NOD mice) reduced systemic and tissue-localized inflammation, prevented β cell death, and delayed T1D onset. Transcriptional phenotyping of pancreatic islets, pancreatic lymph nodes (PLN), and spleen during early disease pathogenesis highlighted a role for TYK2 inhibition in modulating signaling pathways associated with inflammation, translational control, stress signaling, secretory function, immunity, and diabetes. Additionally, TYK2i treatment changed the composition of innate and adaptive immune cell populations in the blood and disease target tissues, resulting in an immune phenotype with a diminished capacity for β cell destruction. Overall, these findings indicate that TYK2i has beneficial effects in both the immune and endocrine compartments in models of T1D, thus supporting a path forward for testing TYK2 inhibitors in human T1D.
Collapse
Affiliation(s)
- Farooq Syed
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Olivia Ballew
- Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Chih-Chun Lee
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jyoti Rana
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Preethi Krishnan
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Angela Castela
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Staci A. Weaver
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Sofia F. Thomaidou
- Department of Cell and Chemical Biology, Leiden University Medical Center, The Netherlands
| | - Stephane Demine
- Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Garrick Chang
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | | | - Maria Ines Alvelos
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Lorella Marselli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Kara Orr
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jamie L. Felton
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jing Liu
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN, USA
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, The Netherlands
| | | | - Decio L. Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Carmella Evans-Molina
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
13
|
Zhang M, Liu Z, Zhu Y, Wu K, Zhou L, Peng Y, Pan J, Chen B, Wang X, Chen S. Associations of genetic variants within TYK2 with pulmonary tuberculosis among Chinese population. Mol Genet Genomic Med 2024; 12:e2386. [PMID: 38337161 PMCID: PMC10858315 DOI: 10.1002/mgg3.2386] [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: 10/11/2023] [Revised: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Pulmonary tuberculosis (PTB) is a common infectious disease caused by mycobacterium tuberculosis (MTB) and the present study aims to explore the associations of genetic variants within tyrosine kinases 2 (TYK2) with PTB incidence. METHODS A population-based case control study including 168 smear-positive PTB cases and 251 controls was conducted. Five single nucleotide polymorphisms (SNPs) including rs280520, rs91755, rs2304256, rs12720270, rs280519 located within TYK2 gene were selected and MassARRAY® MALDI-TOF system was employed for genotyping. SPSS 19.0 was adopted for statistical analysis, non-conditional logistic regression was conducted. Odds ratios (ORs) and 95% confidence intervals (95% CIs) were computed to estimate their contributions to PTB incidence. RESULTS In the overall study population, rs91755 TT and rs280519 AA genotypes were found to be associated with reduced PTB risk (OR = 0.34, 95% CI: 0.16-0.72; OR = 0.38, 95% CI: 0.18-0.79, respectively). After stratification for sex, we found that among the male population, rs91755TG/TT, rs12720270AG/GG and rs280519AG/AA genotypes were associated with reduced PTB risk (OR = 0.41, 95% CI: 0.21-0.80; OR = 0.44, 95% CI: 0.21-0.94; OR = 0.42, 95% CI: 0.21-0.82, respectively). After stratification for age, we found that among those aged <60 years, rs91755TT and rs280519AA genotype were associated with reduced PTB risk (OR = 0.29, 95% CI: 0.09-0.90; OR = 0.34, 95% CI: 0.11-1.08, respectively); while rs2304256AC/AA genotype was associated with increased PTB risk (OR = 2.68, 95% CI: 1.05-6.85). Haplotype analysis revealed that AGAAG and ATCGA (Combined with rs280520, rs91755, rs2304256, rs12720270 and rs280519) were associated with increased (OR = 1.54, 95% CI: 1.01-2.37) and decreased PTB risk (OR = 0.70, 95% CI: 0.52-0.94), respectively. CONCLUSIONS The genetic variants located within TYK2 including rs91755, rs12720270 and rs280519 were found to be associated with modified PTB risk and the SNPs had potential to be the biomarkers to predict PTB incidence risk.
Collapse
Affiliation(s)
- Mingwu Zhang
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| | - Zhengwei Liu
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| | - Yelei Zhu
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| | - Kunyang Wu
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| | - Lin Zhou
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| | - Ying Peng
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| | - Junhang Pan
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| | - Bin Chen
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| | - Xiaomeng Wang
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| | - Songhua Chen
- Tuberculosis Control and PreventionZhejiang Provincial Center for Disease Control and PreventionHangzhou CityChina
| |
Collapse
|
14
|
Cheraghi Z, Ziai SA, Fazeli Z, Gheisoori A. Unveiling sex-based impact of TYK2 rs2304256 polymorphism on interferon beta-1alpha responsiveness in COVID-19 patients. GENE REPORTS 2023; 33:101846. [DOI: 10.1016/j.genrep.2023.101846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
15
|
Jensen LT, Attfield KE, Feldmann M, Fugger L. Allosteric TYK2 inhibition: redefining autoimmune disease therapy beyond JAK1-3 inhibitors. EBioMedicine 2023; 97:104840. [PMID: 37863021 PMCID: PMC10589750 DOI: 10.1016/j.ebiom.2023.104840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/28/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023] Open
Abstract
JAK inhibitors impact multiple cytokine pathways simultaneously, enabling high efficacy in treating complex diseases such as cancers and immune-mediated disorders. However, their broad reach also poses safety concerns, which have fuelled a demand for increasingly selective JAK inhibitors. Deucravacitinib, a first-in-class allosteric TYK2 inhibitor, represents a remarkable advancement in the field. Rather than competing at kinase domain catalytic sites as classical JAK1-3 inhibitors, deucravacitinib targets the regulatory pseudokinase domain of TYK2. It strikingly mirrors the functional effect of an evolutionary conserved naturally occurring TYK2 variant, P1104A, known to protect against multiple autoimmune diseases yet provide sufficient TYK2-mediated cytokine signalling required to prevent immune deficiency. The unprecedentedly high functional selectivity and efficacy-safety profile of deucravacitinib, initially demonstrated in psoriasis, combined with genetic support, and promising outcomes in early SLE clinical trials make this inhibitor ripe for exploration in other autoimmune diseases for which better, safe, and efficacious treatments are urgently needed.
Collapse
Affiliation(s)
- Lise Torp Jensen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Kathrine E Attfield
- Nuffield Department of Clinical Neurosciences, Oxford Centre for Neuroinflammation, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Marc Feldmann
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, The Kennedy Institute for Rheumatology, Botnar Research Institute, University of Oxford, Oxford OX3 7LD, UK
| | - Lars Fugger
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus 8200, Denmark; Nuffield Department of Clinical Neurosciences, Oxford Centre for Neuroinflammation, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK; MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
16
|
Sun Q, Rowland B, Wang W, Miller-Fleming TW, Cox N, Graff M, Faucon A, Shuey MM, Blue EE, Auer P, Li Y, Sankaran VG, Reiner AP, Raffield LM. Genetic examination of hematological parameters in SARS-CoV-2 infection and COVID-19. Blood Cells Mol Dis 2023; 103:102782. [PMID: 37558590 PMCID: PMC10507673 DOI: 10.1016/j.bcmd.2023.102782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023]
Abstract
People hospitalized with COVID-19 often exhibit altered hematological traits associated with disease prognosis (e.g., lower lymphocyte and platelet counts). We investigated whether inter-individual variability in baseline hematological traits influences risk of acute SARS-CoV-2 infection or progression to severe COVID-19. We report inconsistent associations between blood cell traits with incident SARS-CoV-2 infection and severe COVID-19 in UK Biobank and the Vanderbilt University Medical Center Synthetic Derivative (VUMC SD). Since genetically determined blood cell measures better represent cell abundance across the lifecourse, we also assessed the shared genetic architecture of baseline blood cell traits on COVID-19 related outcomes by Mendelian randomization (MR) analyses. We found significant relationships between COVID-19 severity and mean sphered cell volume after adjusting for multiple testing. However, MR results differed significantly across different freezes of COVID-19 summary statistics and genetic correlation between these traits was modest (0.1), decreasing our confidence in these results. We observed overlapping genetic association signals between other hematological and COVID-19 traits at specific loci such as MAPT and TYK2. In conclusion, we did not find convincing evidence of relationships between the genetic architecture of blood cell traits and either SARS-CoV-2 infection or COVID-19 hospitalization, though we do see evidence of shared signals at specific loci.
Collapse
Affiliation(s)
- Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Bryce Rowland
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Wanjiang Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Tyne W Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Nancy Cox
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Misa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Annika Faucon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Megan M Shuey
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Elizabeth E Blue
- Department of Medicine, Division of Medical Genetics, University of Washington, Brotman Baty Institute for Precision Medicine, Seattle, WA, United States
| | - Paul Auer
- Division of Biostatistics, Institute for Health and Equity, Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
17
|
Yudhani RD, Pakha DN, Suyatmi S, Irham LM. Identifying pathogenic variants related to systemic lupus erythematosus by integrating genomic databases and a bioinformatic approach. Genomics Inform 2023; 21:e37. [PMID: 37813633 PMCID: PMC10584638 DOI: 10.5808/gi.23002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/15/2023] [Accepted: 08/09/2023] [Indexed: 10/11/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an inflammatory-autoimmune disease with a complex multi-organ pathogenesis, and it is known to be associated with significant morbidity and mortality. Various genetic, immunological, endocrine, and environmental factors contribute to SLE. Genomic variants have been identified as potential contributors to SLE susceptibility across multiple continents. However, the specific pathogenic variants that drive SLE remain largely undefined. In this study, we sought to identify these pathogenic variants across various continents using genomic and bioinformatic-based methodologies. We found that the variants rs35677470, rs34536443, rs17849502, and rs13306575 are likely damaging in SLE. Furthermore, these four variants appear to affect the gene expression of NCF2, TYK2, and DNASE1L3 in whole blood tissue. Our findings suggest that these genomic variants warrant further research for validation in functional studies and clinical trials involving SLE patients. We conclude that the integration of genomic and bioinformatic-based databases could enhance our understanding of disease susceptibility, including that of SLE.
Collapse
Affiliation(s)
- Ratih Dewi Yudhani
- Department of Pharmacology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta 57126, Indonesia
| | - Dyonisa Nasirochmi Pakha
- Department of Pharmacology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta 57126, Indonesia
| | - Suyatmi Suyatmi
- Department of Histology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta 57126, Indonesia
| | | |
Collapse
|
18
|
Mohammadi B, Dua K, Saghafi M, Singh SK, Heydarifard Z, Zandi M. COVID-19-induced autoimmune thyroiditis: Exploring molecular mechanisms. J Med Virol 2023; 95:e29001. [PMID: 37515444 DOI: 10.1002/jmv.29001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/30/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) damages multiple organs, including the thyroid, by direct invasion and cell entry via angiotensin-converting enzyme 2 or indirectly by promoting excessive inflammation in the body. The immune system is a critical factor in antiviral immunity and disease progression. In the context of SARS-CoV-2 infection, the immune system may become overly activated, resulting in a shift from regulatory to effector responses, which may subsequently promote the development and progression of autoimmune diseases. The incidence of autoimmune thyroid diseases, such as subacute thyroiditis, Graves' disease, and Hashimoto's thyroiditis, increases in individuals with COVID-19 infection. This phenomenon may be attributed to aberrant responses of T-cell subtypes, the presence of autoantibodies, impaired regulatory cell function, and excessive production of inflammatory cytokines, namely interleukin (IL)-6, IL-1β, interferon-γ, and tumor necrosis factor-α. Therefore, insights into the immune responses involved in the development of autoimmune thyroid disease according to COVID-19 can help identify potential therapeutic approaches and guide the development of effective interventions to alleviate patients' symptoms.
Collapse
Affiliation(s)
- Bita Mohammadi
- Department of Immunology, Mashhad University of Medical Sciences, Mashhad, Iran
- Innovated Medical Research Center, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, Australia
- Faculty of Health, Australian Research Center in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Mohammadreza Saghafi
- Department of Immunology, Mashhad University of Medical Sciences, Mashhad, Iran
- Innovated Medical Research Center, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Center in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Zahra Heydarifard
- Department of Virology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
- School of Medicine, Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Dieter C, de Almeida Brondani L, Lemos NE, Schaeffer AF, Zanotto C, Ramos DT, Girardi E, Pellenz FM, Camargo JL, Moresco KS, da Silva LL, Aubin MR, de Oliveira MS, Rech TH, Canani LH, Gerchman F, Leitão CB, Crispim D. Polymorphisms in ACE1, TMPRSS2, IFIH1, IFNAR2, and TYK2 Genes Are Associated with Worse Clinical Outcomes in COVID-19. Genes (Basel) 2022; 14:genes14010029. [PMID: 36672770 PMCID: PMC9858252 DOI: 10.3390/genes14010029] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/29/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022] Open
Abstract
Although advanced age, male sex, and some comorbidities impact the clinical course of COVID-19, these factors only partially explain the inter-individual variability in disease severity. Some studies have shown that genetic polymorphisms contribute to COVID-19 severity; however, the results are inconclusive. Thus, we investigated the association between polymorphisms in ACE1, ACE2, DPP9, IFIH1, IFNAR2, IFNL4, TLR3, TMPRSS2, and TYK2 and the clinical course of COVID-19. A total of 694 patients with COVID-19 were categorized as: (1) ward inpatients (moderate symptoms) or patients admitted at the intensive care unit (ICU; severe symptoms); and (2) survivors or non-survivors. In females, the rs1990760/IFIH1 T/T genotype was associated with risk of ICU admission and death. Moreover, the rs1799752/ACE1 Ins and rs12329760/TMPRSS2 T alleles were associated with risk of ICU admission. In non-white patients, the rs2236757/IFNAR2 A/A genotype was associated with risk of ICU admission, while the rs1799752/ACE1 Ins/Ins genotype, rs2236757/IFNAR2 A/A genotype, and rs12329760/TMPRSS2 T allele were associated with risk of death. Moreover, some of the analyzed polymorphisms interact in the risk of worse COVID-19 outcomes. In conclusion, this study shows an association of rs1799752/ACE1, rs1990760/IFIH1, rs2236757/IFNAR2, rs12329760/TMPRSS2, and rs2304256/TYK2 polymorphisms with worse COVID-19 outcomes, especially among female and non-white patients.
Collapse
Affiliation(s)
- Cristine Dieter
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Leticia de Almeida Brondani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Natália Emerim Lemos
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Ariell Freires Schaeffer
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Caroline Zanotto
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Denise Taurino Ramos
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Eliandra Girardi
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Felipe Mateus Pellenz
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Joiza Lins Camargo
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Karla Suzana Moresco
- Campus Realeza, Universidade Federal da Fronteira Sul, Realeza 85770-000, PR, Brazil
| | - Lucas Lima da Silva
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Mariana Rauback Aubin
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Mayara Souza de Oliveira
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Tatiana Helena Rech
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Luís Henrique Canani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Fernando Gerchman
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Cristiane Bauermann Leitão
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Daisy Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Correspondence:
| |
Collapse
|
20
|
Tziastoudi M, Cholevas C, Stefanidis I, Theoharides TC. Genetics of COVID-19 and myalgic encephalomyelitis/chronic fatigue syndrome: a systematic review. Ann Clin Transl Neurol 2022; 9:1838-1857. [PMID: 36204816 PMCID: PMC9639636 DOI: 10.1002/acn3.51631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 01/08/2023] Open
Abstract
COVID‐19 and ME/CFS present with some similar symptoms, especially physical and mental fatigue. In order to understand the basis of these similarities and the possibility of underlying common genetic components, we performed a systematic review of all published genetic association and cohort studies regarding COVID‐19 and ME/CFS and extracted the genes along with the genetic variants investigated. We then performed gene ontology and pathway analysis of those genes that gave significant results in the individual studies to yield functional annotations of the studied genes using protein analysis through evolutionary relationships (PANTHER) VERSION 17.0 software. Finally, we identified the common genetic components of these two conditions. Seventy‐one studies for COVID‐19 and 26 studies for ME/CFS were included in the systematic review in which the expression of 97 genes for COVID‐19 and 429 genes for ME/CFS were significantly affected. We found that ACE, HLA‐A, HLA‐C, HLA‐DQA1, HLA‐DRB1, and TYK2 are the common genes that gave significant results. The findings of the pathway analysis highlight the contribution of inflammation mediated by chemokine and cytokine signaling pathways, and the T cell activation and Toll receptor signaling pathways. Protein class analysis revealed the contribution of defense/immunity proteins, as well as protein‐modifying enzymes. Our results suggest that the pathogenesis of both syndromes could involve some immune dysfunction.
Collapse
Affiliation(s)
- Maria Tziastoudi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Christos Cholevas
- First Department of Ophthalmology, Faculty of Health Sciences, Aristotle University, AHEPA Hospital, Thessaloniki, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Theoharis C Theoharides
- Institute of Neuro-Immune Medicine, Nova Southeastern University, Clearwater, FL, USA.,Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA.,Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Regulation of activated T cell survival in rheumatic autoimmune diseases. Nat Rev Rheumatol 2022; 18:232-244. [PMID: 35075294 DOI: 10.1038/s41584-021-00741-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2021] [Indexed: 12/29/2022]
Abstract
Adaptive immune responses rely on the proliferation of T lymphocytes able to recognize and eliminate pathogens. The magnitude and duration of the expansion of activated T cell clones are finely regulated to minimize immunopathology and avoid autoimmunity. In patients with rheumatic autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis, activated lymphocytes survive and exert effector functions for prolonged periods, defying the mechanisms that normally curb their capacities during acute and chronic infections. Here, we review the molecular mechanisms that limit the duration of immune responses in health and discuss the factors that alter such regulation in the setting of systemic lupus erythematosus and rheumatoid arthritis. We highlight defects that could contribute to the development and progression of autoimmune disease and describe how chronic inflammation can alter the regulation of activated lymphocyte survival, promoting its perpetuation. These concepts might contribute to the understanding of the mechanisms that underlie the chronicity of inflammation in the context of autoimmunity.
Collapse
|
22
|
Rowland B, Sun Q, Wang W, Miller-Fleming T, Cox N, Graff M, Faucon A, Shuey MM, Blue EE, Auer P, Li Y, Sankaran VG, Reiner AP, Raffield LM. Genetic Examination of Hematological Parameters in SARS-CoV-2 Infection and COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.02.28.22271562. [PMID: 35262092 PMCID: PMC8902884 DOI: 10.1101/2022.02.28.22271562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background People hospitalized with COVID-19 often exhibit hematological alterations, such as lower lymphocyte and platelet counts, which have been reported to associate with disease prognosis. It is unclear whether inter-individual variability in baseline hematological parameters prior to acute infection influences risk of SARS-CoV-2 infection and progression to severe COVID-19. Methods We assessed the association of blood cell counts and indices with incident SARS-CoV-2 infection and severe COVID-19 in UK Biobank and the Vanderbilt University Medical Center Synthetic Derivative (VUMC SD). Since genetically determined blood cell measures better represent cell abundance across the lifecourse, we used summary statistics from genome-wide association studies to assess the shared genetic architecture of baseline blood cell counts and indices on COVID-19 outcomes. Results We observed inconsistent associations between measured blood cell indices and both SARS-CoV-2 infection and COVID-19 hospitalization in UK Biobank and VUMC SD. In Mendelian randomization analyses using genetic summary statistics, no putative causal relationships were identified between COVID-19 related outcomes and hematological indices after adjusting for multiple testing. We observed overlapping genetic association signals between hematological parameters and COVID-19 traits. For example, we observed overlap between infection susceptibility-associated variants at PPP1R15A and red blood cell parameters, and between disease severity-associated variants at TYK2 and lymphocyte and platelet phenotypes. Conclusions We did not find convincing evidence of a relationship between baseline hematological parameters and susceptibility to SARS-CoV-2 infection or COVID-19 severity, though this relationship should be re-examined as larger and better-powered genetic analyses of SARS-CoV-2 infection and severe COVID-19 become available.
Collapse
Affiliation(s)
- Bryce Rowland
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Wanjiang Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Tyne Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Nancy Cox
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Misa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Annika Faucon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Megan M. Shuey
- Department of Medicine Vanderbilt University Medical Center Nashville, TN
| | - Elizabeth E. Blue
- Department of Medical Genetics, University of Washington, Seattle, WA
| | - Paul Auer
- Division of Biostatistics, Institute for Health and Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Vijay G. Sankaran
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
23
|
Muromoto R, Shimoda K, Oritani K, Matsuda T. Therapeutic Advantage of Tyk2 Inhibition for Treating Autoimmune and Chronic Inflammatory Diseases. Biol Pharm Bull 2021; 44:1585-1592. [PMID: 34719635 DOI: 10.1248/bpb.b21-00609] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tyrosine kinase 2 (Tyk2) is a member of the Janus family of protein tyrosine kinases (Jaks). Tyk2 associates with interferon (IFN)-α, IFN-β, interleukin (IL)-6, IL-10, IL-12, and IL-23 receptors and mediates their downstream signaling pathways. Based on our data using Tyk2-deficient mice and cells, Tyk2 plays crucial roles in the differentiation, maintenance, and function of T helper 1 (Th1) and Th17 cells, and its dysregulation may promote autoimmune and/or inflammatory diseases. IFN-α-induced growth inhibition of B lymphocyte progenitors is dependent on Tyk2-mediated signals to regulate death-associated protein (Daxx) nuclear localization and Daxx-promyelocytic leukemia protein interactions. Tyk2-deficient mice show impaired constitutive production of type I IFNs by macrophages under steady-state conditions. When heat-killed Cutibacterium acnes is injected intraperitoneally, Tyk2-deficient mice show less granuloma formation through enhanced prostaglandin E2 and protein kinase A activities, leading to high IL-10 production by macrophages. Thus, Tyk2 is widely involved in the immune and inflammatory response at multiple events; therefore, Tyk2 is likely to be a suitable target for treating patients with autoimmune and/or chronic inflammatory diseases. Clinical trials of Tyk2 inhibitors have shown higher response rates and improved tolerability in the treatment of patients with psoriasis and inflammatory bowel diseases. Taken together, Tyk2 inhibition has great potential for clinical application in the management of a variety of diseases.
Collapse
Affiliation(s)
- Ryuta Muromoto
- Department of Immunology, Graduate School of Pharmaceutical Sciences Hokkaido University
| | - Kazuya Shimoda
- Department of Internal Medicine II, Faculty of Medicine, University of Miyazaki
| | - Kenji Oritani
- Department of Hematology, International University of Health and Welfare
| | - Tadashi Matsuda
- Department of Immunology, Graduate School of Pharmaceutical Sciences Hokkaido University
| |
Collapse
|