1
|
Li H, Zhang X, Zhaxi Y, Pan C, Zhang Z, Pan J, Shahzad K, Sun F, Zhen Y, Jinmei J, Zhao W, Song T. Integrative multi-omics analysis reveals liver-gut axis adaptation in high-altitude goats. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101422. [PMID: 39842302 DOI: 10.1016/j.cbd.2025.101422] [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: 10/03/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
The liver-gut axis is an important regulatory axis for the host's metabolic functions. The study of liver gene expression, changes in metabolic products and the regulation of gut microbial communities in plateau animals under harsh environments can reveal the mechanisms by which Tibetan goats adapt to the plateau environment. This study employs transcriptome, metabolome and metagenomic analyses to reveal the differences in genes, metabolism, and gut microbiota between Jianzhou big-eared goats (JBG) and Xizang cashmere goats (TCG), which is of significant importance for improving survival models of high-altitude ruminants. The results showed that there were 553 DEGs in the liver of JBG and TCG. Hepatic metabolomic analysis revealed significant differences in metabolic activity between the JBG and TCG groups, with notable increases in glycerophospholipid and retinol metabolic pathways. The gut microbiota, including Andreesenia, Dielma, Oscillibacter, Agrobacterium, Hyella and Thermosinus, interact with liver metabolites and can regulate the high-altitude adaptability of goats. This study reveals that TCG enhance immune regulation and energy utilization efficiency by regulating liver gene expression, modulating metabolic pathways, and improving gut microbiota, thereby helping TCG maintain healthy survival capabilities in hypoxic and high-radiation environments.
Collapse
Affiliation(s)
- Haiyan Li
- College of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China; Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, Xizang 850009, China
| | - Xin Zhang
- College of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| | - Yangzong Zhaxi
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, Xizang 850009, China; Key Laboratory of Animal Genetics and Breeding on Xizang Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, Xizang 850009, China
| | - Cheng Pan
- College of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| | - Zhenzhen Zhang
- College of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| | - Junru Pan
- College of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| | - Khuram Shahzad
- Department of Biosciences, COMSATS University Islamabad, Park Road, Islamabad 45550, Pakistan
| | - Fengbo Sun
- Xizang Animal Husbandry Station, Lhasa, Xizang 850000, China
| | - Yang Zhen
- Xizang Animal Husbandry Station, Lhasa, Xizang 850000, China
| | - Jiacuo Jinmei
- Xizang Animal Husbandry Station, Lhasa, Xizang 850000, China
| | - Wangsheng Zhao
- College of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China.
| | - Tianzeng Song
- Institute of Animal Science, Xizang Academy of Agricultural and Animal Husbandry Science, Lhasa, Xizang 850009, China; Key Laboratory of Animal Genetics and Breeding on Xizang Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, Xizang 850009, China.
| |
Collapse
|
2
|
Plug A, Barenbrug L, Moerings BG, de Jong EM, van der Molen RG. Understanding the role of immune-mediated inflammatory disease related cytokines interleukin 17 and 23 in pregnancy: A systematic review. J Transl Autoimmun 2025; 10:100279. [PMID: 40035074 PMCID: PMC11874717 DOI: 10.1016/j.jtauto.2025.100279] [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: 12/11/2024] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
Background Pregnancy requires a careful immune balance between tolerance for the semi-allogenic fetus and protection against pathogens. Women with immune-mediated inflammatory diseases (IMIDs), where the interleukin (IL)-23/IL-17 axis plays an important role, often experience changes in disease severity during pregnancy. These changes and the association between disease flares and pregnancy complications, suggests a role for IL-17 and IL-23 in pregnancy. Methods We systemically searched PubMed, EMBASE, and Web of Science (March 2024), to assess the role of IL-17 and IL-23 in pregnancy-related in vitro assays, animal or human studies. Results Eighty articles (8 in vitro, 11 animal and 61 human studies) were included. Seventy-one studies reported on IL-17 and 16 studies on IL-23. In vitro trophoblast proliferation, migration and invasion was increased in the presence of IL-17, but impaired with IL-23. IL-17 levels were increased in animal models for pregnancy complications. In humans, IL-17 levels seemed to be increased in pregnant women versus non-pregnant women. Additionally, elevated IL-17 levels were associated with pregnancy complications. Although similar trends were found for IL-23, data were limited. Conclusions We identified a large, but heterogenic, body of evidence for a significant role of IL-17 in all stages of pregnancy: while an excessive increase seemed to be associated with complications. The limited number of studies prevents firm conclusions on the role of IL-23. Future research is needed to find biomarkers for patients with IMIDs to predict the effect of possible disease flares on pregnancy, and the effect of therapeutic inhibition of IL-17 or IL-23.
Collapse
Affiliation(s)
- Aniek Plug
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center (Radboudumc), Nijmegen, the Netherlands
| | - Liana Barenbrug
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, the Netherlands
| | - Bart G.J. Moerings
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center (Radboudumc), Nijmegen, the Netherlands
| | - Elke M.G. de Jong
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, the Netherlands
| | - Renate G. van der Molen
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center (Radboudumc), Nijmegen, the Netherlands
| |
Collapse
|
3
|
Yang Z, Jiang Y, Ma J, Wang L, Han S, Huda N, Kusumanchi P, Gao H, Thoudam T, Sun Z, Liangpunsakul S. LncRNA H19 promoted alcohol-associated liver disease through dysregulation of alternative splicing and methionine metabolism. Hepatology 2025; 81:1485-1500. [PMID: 39364651 DOI: 10.1097/hep.0000000000001078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/30/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND AND AIMS Long noncoding RNAs constitute a significant portion of the human genome. Among these, lncRNA H19, initially identified for its high expression during fetal development followed by a decline in the liver postnatally, re-emerges in various liver diseases. However, its specific role in alcohol-associated liver disease (ALD) remains unclear. APPROACH AND RESULTS Elevated H19 levels were detected in peripheral blood and livers of patients with alcohol-associated cirrhosis and hepatitis, as well as in livers of ethanol-fed mice. Hepatic overexpression of H19 exacerbated ethanol-induced liver steatosis and injury. Metabolomics analysis revealed decreased methionine levels in H19-overexpressed mouse livers, attributable to H19-mediated inhibition of betaine homocysteine methyltransferase (BHMT), a crucial enzyme in methionine synthesis. H19 regulated BHMT alternative splicing through polypyrimidine tract-binding protein 1 (PTBP1), resulting in a reduced Bhmt protein-coding variant. The maternally specific knockout of H19 ( H19Mat+/- ) or liver-specific knockout of the H19 differentially methylated domain ( H19DMDHep-/- ) in ethanol-fed mice upregulated BHMT expression and ameliorated hepatic steatosis. Furthermore, BHMT restoration counteracted H19-induced ethanol-mediated hepatic steatosis. CONCLUSIONS This study identifies a novel mechanism whereby H19, via PTBP1-mediated BHMT regulation, influences methionine metabolism in ALD. Targeting the H19-PTBP1-BHMT pathway may offer new therapeutic avenues for ALD.
Collapse
Affiliation(s)
- Zhihong Yang
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yanchao Jiang
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jing Ma
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Li Wang
- Independent Researcher, Tucson, Arizona, USA
| | - Sen Han
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nazmul Huda
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Praveen Kusumanchi
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hui Gao
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Themis Thoudam
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Suthat Liangpunsakul
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
4
|
Li L, Liu J, Lu J, Wu J, Zhang X, Ma T, Wu X, Zhu Q, Chen Z, Tai Z. Interventions in cytokine signaling: novel horizons for psoriasis treatment. Front Immunol 2025; 16:1573905. [PMID: 40303401 PMCID: PMC12037536 DOI: 10.3389/fimmu.2025.1573905] [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: 02/10/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Intricate interactions between immune cells and cytokines define psoriasis, a chronic inflammatory skin condition that is immunological-mediated. Cytokines, including interleukins (ILs), interferons (IFNs), tumor necrosis factors (TNFs), chemokines, and transforming growth factor-β (TGF-β), are essential for controlling cellular activity and immunological responses, maintaining homeostasis and contributing to the pathogenesis of psoriasis. These molecules modulate the immune microenvironment by either promoting or suppressing inflammation, which significantly impacts therapeutic outcomes. Recent research indicates that treatment strategies targeting cytokines and chemokines have significant potential, offering new approaches for regulating the immune system, inhibiting the progression of psoriasis, and reducing adverse effects of traditional therapies. This review consolidates current knowledge on cytokine and chemokine signaling pathways in psoriasis and examines their significance in treatment. Specific attention is given to cytokines like IL-17, IL-23, and TNF-α, underscoring the necessity for innovative therapies to modulate these pathways and address inflammatory processes. This review emphasizes the principal part of cytokines in the -pathological process of psoriasis and explores the challenges and opportunities they present for therapeutic intervention. Furthermore, we examine recent advancements in targeted therapies, with a particular focus on monoclonal antibodies, in ongoing research and clinical trials.
Collapse
Affiliation(s)
- Lisha Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Jiaye Lu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Junchao Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Tianyou Ma
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Xiying Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Raavi, Koehler AN, Vegas AJ. At The Interface: Small-Molecule Inhibitors of Soluble Cytokines. Chem Rev 2025. [PMID: 40233276 DOI: 10.1021/acs.chemrev.4c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cytokines are crucial regulators of the immune system that orchestrate interactions between cells and, when dysregulated, contribute to the progression of chronic inflammation, cancer, and autoimmunity. Numerous biologic-based clinical agents, mostly monoclonal antibodies, have validated cytokines as important clinical targets and are now part of the standard of care for a number of diseases. These agents, while impactful, still suffer from limitations including a lack of oral bioavailability, high cost of production, and immunogenicity. Small-molecule cytokine inhibitors are attractive alternatives that can address these limitations. Although targeting cytokine-cytokine receptor complexes with small molecules has been a challenging research endeavor, multiple small-molecule inhibitors have now been identified, with a number of them undergoing clinical evaluation. In this review, we highlight the recent advancements in the discovery and development of small-molecule inhibitors targeting soluble cytokines. The strategies for identifying these novel ligands as well as the structural and mechanistic insights into their activity represent important milestones in tackling these challenging and clinically important protein-protein interactions.
Collapse
Affiliation(s)
- Raavi
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Angela N Koehler
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Arturo J Vegas
- Department of Chemistry, Boston University, Boston, Massachusetts 02115, United States
| |
Collapse
|
6
|
Taş Ö, Aydın F, Kuloğlu Z, Kırsaçlıoğlu CT, Bahçeci O, Aydın BÖ, Sarısoy D, Özçakar ZB. Inflammatory bowel disease in paediatric rheumatological diseases. Clin Rheumatol 2025:10.1007/s10067-025-07424-w. [PMID: 40186043 DOI: 10.1007/s10067-025-07424-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
INTRODUCTION Rheumatological diseases (RD) in childhood are systemic diseases that occur on the basis of auto-immunity or inflammation, and they can be accompanied by inflammatory bowel disease (IBD). When there is no knowledge of this association, the treatments applied may not be sufficient and/or treatments given for RD may even lead to aggrevation of IBD findings. Thus, early identification of an association is crucial for the correct management of the diseases. OBJECTIVES The aim of this study is to show the frequency of IBD in patients with RDs. We also aimed to investigate in which cases IBD should be suspected in children with RDs. METHODS Electronic medical records of the patients who were followed up between 2012 and 2024 with a diagnosis of RD in our Paediatric Rheumatology Unit and diagnosed with IBD were reviewed retrospectively. RESULTS Between 2012 and 2024, 20 (3%) of 650 familial Mediterranean fever (FMF) patients, 3 (7.5%) of 40 chronic nonbacterial osteomyelitis (CNO) patients (one of them also had FMF) and 2 (1.2%) of 170 juvenile idiopathic arthritis (JIA) patients were diagnosed with IBD. While 15 (62.5%) of the patients received a RD as the initial diagnosis, 9 (37.5%) of them were initially diagnosed with IBD and then referred to rheumatology for their symptoms. The median age at the diagnosis of RD was 9 years (inter quartile range (IQR), 14.5). The median age at the diagnosis of IBD was 12 years (IQR, 13), and 12 patients (50%) had Crohn's disease (CD), 10 patients (41.6%) ulcerative colitis (UC) and 2 patients (8.4%) undeterminated disease. Although majority of the patients had classical findings of IBD, 4 patients presented with more vague symptoms including treatment-resistant iron deficiency anaemia, perianal abscess, weight loss and growth retardation. CONCLUSIONS RD and IBD share similar pathological pathways and clinical findings, and IBD can accompany to various RD. The diagnosis of IBD should be considered in the presence of rare and atypical symptoms. Furthermore, RD should also be considered in children with IBD who have complex extraintestinal symptoms. Key Points • RD, especially FMF, JIA and CNO, may be associated with IBD. • Paediatric rheumatologists should consider IBD as a potential diagnosis in the presence of atypical findings that may develop during the course of RD. • The co-existence of RD and IBD is important both in terms of disease progression and treatment decisions.
Collapse
Affiliation(s)
- Özen Taş
- Division of Pediatric Rheumatology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Fatma Aydın
- Division of Pediatric Rheumatology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Zarife Kuloğlu
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Ceyda Tuna Kırsaçlıoğlu
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Onur Bahçeci
- Division of Pediatric Rheumatology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Betül Öksüz Aydın
- Division of Pediatric Rheumatology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Doğacan Sarısoy
- Division of Pediatric Rheumatology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Zeynep Birsin Özçakar
- Division of Pediatric Nephrology and Rheumatology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey.
| |
Collapse
|
7
|
Madill E, Galetta K, Opeyemi O, Pua DK, Gandelman S, Chitnis T, Bhattacharyya S. Safety and efficacy of anti-IL-17A use in multiple sclerosis and comorbid rheumatological disease: A multi-center exploratory study. J Clin Neurosci 2025; 136:111211. [PMID: 40174548 DOI: 10.1016/j.jocn.2025.111211] [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: 12/10/2024] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Abstract
Anti-IL-17A antibodies are used in rheumatological conditions. While not approved for multiple sclerosis (MS), anti-IL-17As reduce new gadolinium-enhancing lesions. Optimal treatment for those with MS and comorbid rheumatological conditions remains unclear. We report safety and efficacy outcomes for anti-IL-17A treatment with and without concurrent MS disease modifying therapy (DMT). Patients with MS and anti-IL-17A use were identified using electronic medical records. Primary outcomes were severe infections and markers of immunosuppression. Secondary outcomes were MS relapses and new MRI lesions. Six patients (median age: 50.1) without recent MS disease activity had anti-IL-17A monotherapy exposures (17.4 total patient-years); seven (median age: 48.2) had concurrent MS DMT use (8.8 patient-years), including anti-CD20 treatment in three patients. One patient on anti-IL-17A monotherapy had a serious infection. No patients had new or worsening lymphopenia. Four of six patients on anti-IL-17A monotherapy had new MS disease activity. No relapses or new MRI lesions occurred during concurrent MS DMT use. No significant safety concerns were identified with anti-IL-17A and MS DMT combination therapy, although exposure duration was limited. More MS disease activity was seen with anti-IL-17A monotherapy. Dual therapy with an MS DMT may be reasonable for MS patients who require anti-IL-17A treatment.
Collapse
Affiliation(s)
- Evan Madill
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Kristin Galetta
- Department of Neurology, Stanford University, Stanford, CA, USA
| | | | - Danielle Kei Pua
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Tanuja Chitnis
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Shamik Bhattacharyya
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Nguyen KT, Jaguan D, Goldfarb N. Future Directions and Pipeline Therapies for Hidradenitis Suppurativa. Dermatol Clin 2025; 43:359-371. [PMID: 40023632 DOI: 10.1016/j.det.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Hidradenitis suppurativa (HS) is a painful, autoinflammatory condition with dramaticimpact on quality of life. As of January 1, 2024, there are 25 phase I-III clinical trials for topical and systematic medications in the management of HS. Only three medications, adalimumab, secukinumab and bemikizumab, are currently approved by the United States Food and Drug Administration for the treatment of moderate to severe HS. In addition, 15 nonmedication-based studies are ongoing, including those for various surgical techniques, botulinum toxin, laser and light-based therapies, differing wound care methods, and a variety of complementary and alternative medicine practices for HS.
Collapse
Affiliation(s)
- Kim T Nguyen
- Department of Dermatology, University of Minnesota, Minneapolis, MN, USA; College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Daniella Jaguan
- Georgetown University School of Medicine, Washington, DC, USA
| | - Noah Goldfarb
- Department of Dermatology, University of Minnesota, Minneapolis, MN, USA; Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Departments of Medicine and Dermatology, Minneapolis Veteran Affairs Health Care System, Minneapolis, MN, USA.
| |
Collapse
|
9
|
Géczi D, Klekner Á, Balogh I, Penyige A, Szilágyi M, Virga J, Bakó A, Nagy B, Torner B, Birkó Z. Identification of Deregulated miRNAs and mRNAs Involved in Tumorigenesis and Detection of Glioblastoma Patients Applying Next-Generation RNA Sequencing. Pharmaceuticals (Basel) 2025; 18:431. [PMID: 40143207 PMCID: PMC11944724 DOI: 10.3390/ph18030431] [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: 02/07/2025] [Revised: 03/10/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
(1) Background: Glioblastoma (GBM) is one of the most aggressive brain tumors with a poor prognosis. Therefore, new insights into GBM diagnosis and treatment are required. In addition to differentially expressed mRNAs, miRNAs may have the potential to be applied as diagnostic biomarkers. (2) Methods: In this study, profiling of human miRNAs in combination with mRNAs was performed on total RNA isolated from tissue samples of five control and five GBM patients, using a high-throughput RNA sequencing (RNA-Seq) approach. (3) Results: A total of 35 miRNAs and 365 mRNAs were upregulated, while 82 miRNAs and 1225 mRNAs showed significant downregulation between tissue samples of GBM patients compared to the control samples using the iDEP tool to analyze RNA-Seq data. To validate our results, the expression of five miRNAs (hsa-miR-196a-5p, hsa-miR-21-3p, hsa-miR-10b-3p, hsa-miR-383-5p, and hsa-miR-490-3p) and fourteen mRNAs (E2F2, HOXD13, VEGFA, CDC45, AURKB, HOXC10, MYBL2, FABP6, PRLHR, NEUROD6, CBLN1, HRH3, HCN1, and RELN) was determined by RT-qPCR assay. The miRNet tool was used to build miRNA-target interaction. Furthermore, a protein-protein interaction (PPI) network was created from the miRNA targets by applying the NetworkAnalyst 3.0 tool. Based on the PPI network, a functional enrichment analysis of the target proteins was also carried out. (4) Conclusions: We identified an miRNA panel and several deregulated mRNAs that could play an important role in tumor development and distinguish GBM patients from healthy controls with high sensitivity and specificity using total RNA isolated from tissue samples.
Collapse
Affiliation(s)
- Dóra Géczi
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - Álmos Klekner
- Department of Neurosurgery, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - István Balogh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - András Penyige
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - Melinda Szilágyi
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - József Virga
- Department of Oncology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (J.V.); (A.B.)
| | - Andrea Bakó
- Department of Oncology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (J.V.); (A.B.)
| | - Bálint Nagy
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - Bernadett Torner
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - Zsuzsanna Birkó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| |
Collapse
|
10
|
Lin N, Abbas-Aghababazadeh F, Su J, Wu AJ, Lin C, Shi W, Xu W, Haibe-Kains B, Liu FF, Kwan JYY. Development of Machine Learning Models for Predicting Radiation Dermatitis in Breast Cancer Patients Using Clinical Risk Factors, Patient-Reported Outcomes, and Serum Cytokine Biomarkers. Clin Breast Cancer 2025:S1526-8209(25)00048-5. [PMID: 40155248 DOI: 10.1016/j.clbc.2025.03.002] [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: 12/10/2024] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Radiation dermatitis (RD) is a significant side effect of radiotherapy experienced by breast cancer patients. Severe symptoms include desquamation or ulceration of irradiated skin, which impacts quality of life and increases healthcare costs. Early identification of patients at risk for severe RD can facilitate preventive management and reduce severe symptoms. This study evaluated the utility of subjective and objective factors, such as patient-reported outcomes (PROs) and serum cytokines, for predicting RD in breast cancer patients. The performance of machine learning (ML) and logistic regression-based models were compared. PATIENTS AND METHODS Data from 147 breast cancer patients who underwent radiotherapy was analyzed to develop prognostic models. ML algorithms, including neural networks, random forest, XGBoost, and logistic regression, were employed to predict clinically significant Grade 2+ RD. Clinical factors, PROs, and cytokine biomarkers were incorporated into the risk models. Model performance was evaluated using nested cross-validation with separate loops for hyperparameter tuning and calculating performance metrics. RESULTS Feature selection identified 18 predictors of Grade 2+ RD including smoking, radiotherapy boost, reduced motivation, and the cytokines interleukin-4, interleukin-17, interleukin-1RA, interferon-gamma, and stromal cell-derived factor-1a. Incorporating these predictors, the XGBoost model achieved the highest performance with an area under the curve (AUC) of 0.780 (95% CI: 0.701-0.854). This was not significantly improved over the logistic regression model, which demonstrated an AUC of 0.714 (95% CI: 0.629-0.798). CONCLUSION Clinical risk factors, PROs, and serum cytokine levels provide complementary prognostic information for predicting severe RD in breast cancer patients undergoing radiotherapy. ML and logistic regression models demonstrated comparable performance for predicting clinically significant RD with AUC>0.70.
Collapse
Affiliation(s)
- Neil Lin
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Farnoosh Abbas-Aghababazadeh
- Princess Margaret Bioinformatics and Computational Genomics Laboratory, University Health Network, Toronto, Canada
| | - Jie Su
- Biostatistics Division, Princess Margaret Cancer Centre, Toronto, Canada
| | - Alison J Wu
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Cherie Lin
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Wei Shi
- Research Institute, Princess Margaret Cancer Centre, Toronto, Canada
| | - Wei Xu
- Biostatistics Division, Princess Margaret Cancer Centre, Toronto, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Bioinformatics and Computational Genomics Laboratory, University Health Network, Toronto, Canada; Research Institute, Princess Margaret Cancer Centre, Toronto, Canada; Department of Computer Science, University of Toronto, Toronto, Canada; Ontario Institute for Cancer Research, Toronto, Canada; Vector Institute for Artificial Intelligence, Toronto, Canada; Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Fei-Fei Liu
- Research Institute, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Jennifer Y Y Kwan
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Research Institute, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.
| |
Collapse
|
11
|
Sarrand J, Baglione L, Bouvy C, Soyfoo M. Bimekizumab in the Treatment of Axial Spondyloarthritis and Psoriatic Arthritis: A New Kid on the Block. Int J Mol Sci 2025; 26:2315. [PMID: 40076933 PMCID: PMC11899827 DOI: 10.3390/ijms26052315] [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: 01/30/2025] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
The interleukin (IL)-17 family encompasses six structurally related pro-inflammatory cystine knot proteins, designated as IL-17A to IL-17F. Over the last decades, evidence has pointed to its role as a critical player in the development of inflammatory diseases such as psoriasis (PsO), axial spondyloarthritis (axSpA), and psoriatic arthritis (PsA). More specifically, IL-17A and IL-17F are overexpressed in the skin and synovial tissues of patients with these diseases, and recent studies suggest their involvement in promoting inflammation and tissue damage in axSpA and PsA. Bimekizumab is a monoclonal antibody targeting both IL-17A and IL-17F, playing an important role in the treatment of these diseases. This review details the implications of bimekizumab in the therapeutic armamentarium of axSpA and PsA.
Collapse
Affiliation(s)
| | | | | | - Muhammad Soyfoo
- Department of Rheumatology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Bruxelles, Belgium; (J.S.); (L.B.); (C.B.)
| |
Collapse
|
12
|
Han X, Su X, Che M, Liu L, Nie P, Wang S. Identification and Expression Analyses of IL-17/IL-17R Gene Family in Snakehead ( Channa argus) Following Nocardia seriolae Infection. Genes (Basel) 2025; 16:253. [PMID: 40149405 PMCID: PMC11942210 DOI: 10.3390/genes16030253] [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: 11/30/2024] [Revised: 01/18/2025] [Accepted: 02/20/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES The interleukin 17 (IL-17) family, known for its proinflammatory properties, is important in immune responses against bacterial and fungal infections. To exert its immune function, the IL-17 family typically binds to IL-17 receptor (IL-17R) to facilitate signal transduction. METHODS This study identified, cloned and analyzed seven IL-17 and nine IL-17R family members in snakeheads. RESULTS A duplication event occurred in snakehead IL-17s and IL-17Rs, but bioinformatics analyses indicated that these genes were conserved in both protein domains and evolutionary processes. Tissue distribution analysis revealed that IL-17s/IL-17Rs were widely distributed in the detected tissues, with relatively high expression levels in immune tissues. Upon Nocardia seriolae stimulation, most members were expressed, particularly IL-17C2, IL-17D, IL-17N, IL-17RA1, IL-17RA2, IL-17RC1, and IL-17RE1, which were significantly upregulated in gill and intestine. CONCLUSIONS These results suggested that IL-17s and IL-17Rs played a crucial role in mucosal immunity against bacterial infection, providing insights into immunoprophylactic strategies for bacterial diseases in aquaculture.
Collapse
Affiliation(s)
- Xiufeng Han
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao 266109, China; (X.H.); (X.S.); (M.C.); (L.L.)
| | - Xue Su
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao 266109, China; (X.H.); (X.S.); (M.C.); (L.L.)
| | - Mingyue Che
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao 266109, China; (X.H.); (X.S.); (M.C.); (L.L.)
| | - Lanhao Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao 266109, China; (X.H.); (X.S.); (M.C.); (L.L.)
| | - Pin Nie
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao 266109, China; (X.H.); (X.S.); (M.C.); (L.L.)
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Su Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao 266109, China; (X.H.); (X.S.); (M.C.); (L.L.)
| |
Collapse
|
13
|
Chen C, Khanthiyong B, Thaweetee-Sukjai B, Charoenlappanit S, Roytrakul S, Surit P, Phoungpetchara I, Thanoi S, Reynolds GP, Nudmamud-Thanoi S. Proteomic associations with cognitive variability as measured by the Wisconsin Card Sorting Test in a healthy Thai population: A machine learning approach. PLoS One 2025; 20:e0313365. [PMID: 39977438 PMCID: PMC11841870 DOI: 10.1371/journal.pone.0313365] [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: 10/23/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Inter-individual cognitive variability, influenced by genetic and environmental factors, is crucial for understanding typical cognition and identifying early cognitive disorders. This study investigated the association between serum protein expression profiles and cognitive variability in a healthy Thai population using machine learning algorithms. We included 199 subjects, aged 20 to 70, and measured cognitive performance with the Wisconsin Card Sorting Test. Differentially expressed proteins (DEPs) were identified using label-free proteomics and analyzed with the Linear Model for Microarray Data. We discovered 213 DEPs between lower and higher cognition groups, with 155 upregulated in the lower cognition group and enriched in the IL-17 signaling pathway. Subsequent bioinformatic analysis linked these DEPs to neuroinflammation-related cognitive impairment. A random forest model classified cognitive ability groups with an accuracy of 81.5%, sensitivity of 65%, specificity of 85.9%, and an AUC of 0.79. By targeting a specific Thai cohort, this research provides novel insights into the link between neuroinflammation and cognitive performance, advancing our understanding of cognitive variability, highlighting the role of biological markers in cognitive function, and contributing to developing more accurate machine learning models for diverse populations.
Collapse
Affiliation(s)
- Chen Chen
- Faculty of Medical Science, Medical Science graduate program, Naresuan University, Phitsanulok, Thailand
| | | | | | - Sawanya Charoenlappanit
- National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Sittiruk Roytrakul
- National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Phrutthinun Surit
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Ittipon Phoungpetchara
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Samur Thanoi
- School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Gavin P. Reynolds
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - Sutisa Nudmamud-Thanoi
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
14
|
Begagic E, Vranic S, Sominanda A. The role of interleukin 17 in cancer: a systematic review. Carcinogenesis 2025; 46:bgae079. [PMID: 39673782 DOI: 10.1093/carcin/bgae079] [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: 09/07/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 12/16/2024] Open
Abstract
Interleukin 17 (IL17) is a cytokine involved in immune regulation and has been increasingly recognized for its role in cancer progression. This systematic review aims to integrate data on IL17's role in various tumors to better understand its implications for cancer prognosis and treatment. The review included 105 studies (27.6% experimental and 72.4% clinical). Clinical studies involved 9266 patients: 31.2% males, 60.0% females, and 8.8% with undefined gender. IL17A and IL17 were the most studied subtypes (36.2% and 33.3%, respectively). Breast cancer (26.7%), colorectal carcinoma (13.3%), and hematologic malignancies (10.5%) were the most researched neoplasms. IL17A promoted tumor growth in breast cancer and correlated with poor outcomes in colorectal, breast, and lung cancers. IL17 also played a significant role in immune modulation in gliomas and other tumors. IL17A significantly influences tumor growth and prognosis across various cancers, with notable roles in immune modulation and poor outcomes in multiple cancer types.
Collapse
Affiliation(s)
- Emir Begagic
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina
| | - Semir Vranic
- Department of Pathology, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Ajith Sominanda
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| |
Collapse
|
15
|
Ferreira B, Ferreira C, Martins C, Nunes R, das Neves J, Leite-Pereira C, Sarmento B. Establishment of a 3D multi-layered in vitro model of inflammatory bowel disease. J Control Release 2025; 377:675-688. [PMID: 39617170 DOI: 10.1016/j.jconrel.2024.11.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/05/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024]
Abstract
Crohn's Disease and Ulcerative Colitis, the main types of Inflammatory Bowel Disease (IBD), are life-threatening gastrointestinal disorders with no definitive cure. The establishment of biorelevant in vitro models that closely recapitulate the IBD microenvironment is of utmost importance to validate newly developed IBD therapies. To address the existing flaws in the current representation of the IBD microenvironment, we propose a novel three-dimensional (3D) in vitro model comprising a multi-layered gastrointestinal tissue with functional immune responses under inflammatory conditions. The multi-layered architecture consists of a lamina propria-like hydrogel with human intestinal fibroblasts (HIF), supporting an epithelial layer composed of Caco-2 and HT29-MTX cells, along with an endothelial layer surrogating the absorptive capillary network. A collagen-alginate composite matrix was optimized for the lamina propria-like hydrogel, preserving HIF metabolic activity and morphology over time. To achieve immune competence, pre-differentiated THP-1-derived macrophages were incorporated into the epithelial barrier. Inflammation was induced through the optimization of an inflammatory cocktail consisting of E. coli O111:B4 lipopolysaccharide combined with a specialized cytokine array (tumor necrosis factor-α, interferon-γ, and interleukin-1β). This inflammation-inducing stimulus led to a significant upregulation of pro-inflammatory cytokines commonly associated with IBD onset, including CCL20, IL-6, CXCL9 and CXCL10. Altogether, this 3D in vitro model has the potential to accelerate the drug development pipeline by providing reliable permeability and efficacy outputs for emerging therapies, reducing unnecessary animal experiments. Moreover, it offers a valuable in vitro platform for studying IBD pathophysiology and cell interplay dynamics.
Collapse
Affiliation(s)
- Bárbara Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Cecília Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; FCUP - Faculdade de Ciências da Universidade do Porto, Universidade do Porto, Porto, Portugal
| | - Cláudia Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Rute Nunes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - José das Neves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Leite-Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
16
|
Park JW, Rarison RH, Truong VL, Jeong WS. Exploring the Therapeutic Potentials and Molecular Mechanisms of Coscinium fenestratum Alkaloids in Ulcerative Colitis: An Integrative Network Pharmacology and Molecular Docking Approach. Prev Nutr Food Sci 2024; 29:441-453. [PMID: 39759821 PMCID: PMC11699582 DOI: 10.3746/pnf.2024.29.4.441] [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: 10/29/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 01/07/2025] Open
Abstract
Coscinium fenestratum, a medicinal plant traditionally used in Southeast Asia, exerts protective effects against various inflammatory diseases, primarily due to its rich alkaloid content. Despite substantial evidence supporting its anti-inflammatory properties, the biological activities of C. fenestratum are unclear. This study aimed to elucidate anticolitis mechanisms of C. fenestratum alkaloids (CFAs) using an integrative approach of network pharmacology and molecular docking analyses. Key active alkaloids and core target genes were identified through pharmacological and protein-protein interaction networks. The core targets were enriched in the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathways to determine the functional properties of active CFA. Finally, the binding affinity of the key compounds with the core targets was determined using molecular docking. The results showed that 11 active CFAs interactively interfered with 10 hub genes related to ulcerative colitis, including prostaglandin-endoperoxide synthase 2 (PTGS2), selectin E (SELE), kinase insert domain receptor (KDR), fms-related receptor tyrosine kinase 1 (FLT1), intracellular adhesion molecule 1 (ICAM1), C-X-C motif chemokine receptor 4 (CXCR4), hypoxia-inducible factor-1 (HIF1A), matrix metalloproteinase (MMP)-2, MMP3, and MMP9, which were functionally involved in the immunological response, tumor necrosis factor signaling pathway, and interleukin-17 signaling pathway. The molecular docking results indicated that CFA compounds had a strong binding affinity for the hub genes. The findings reveal, for the first time, a therapeutic role of CFA in alleviating ulcerative colitis through its predicted interactions with relevant biological targets.
Collapse
Affiliation(s)
- Ji-Won Park
- School of Food Science & Biotechnology, Kyungpook National University, Daegu 41566, Korea
| | | | - Van-Long Truong
- Food and Bio-industry Research Institute, School of Food Science & Biotechnology College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Woo-Sik Jeong
- School of Food Science & Biotechnology, Kyungpook National University, Daegu 41566, Korea
- Food and Bio-industry Research Institute, School of Food Science & Biotechnology College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
17
|
Zhang C, Weng Y, Wang H, Zhan S, Li C, Zheng D, Lin Q. A synergistic effect of triptolide and curcumin on rheumatoid arthritis by improving cell proliferation and inducing cell apoptosis via inhibition of the IL-17/NF-κB signaling pathway. Int Immunopharmacol 2024; 142:112953. [PMID: 39226828 DOI: 10.1016/j.intimp.2024.112953] [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: 02/01/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 09/05/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic, progressive, systemic autoimmune disease. While triptolide (TPL) and curcumin (CUR) are known to have multiple beneficial effects on RA, the combined effect of TPL and CUR remains unexplored. This study aimed to investigate their synergistic effect on cell proliferation and apoptosis via the IL-17/NF-κB signaling pathway. The collagen-induced arthritis (CIA) rat model was established, showing severe joint and synovial damage compared to normal rats. Treatment with TPL and CUR reduced the severity of RA in the CIA rat model and alleviated serum inflammatory cytokines, such as rheumatoid factor, IL-17, TNF-α, IL-1β, and IL-6. The elevated levels of IL-17 and NF-κB in CIA rats were also inhibited, and the resistant apoptosis was aggravated by TPL and CUR. In vitro, the improvement of cell proliferation and induction of apoptosis were observed in LPS-stimulated MH7A cells treated with TPL and CUR, associated with the inhibition of the IL-17/NF-κB signaling pathway. Taken together, a synergistic effect of TPL and CUR on RA may involve relieving symptoms, improving excessive proliferation, inducing apoptosis resistance, and inhibiting the IL-17/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chaofeng Zhang
- Department of Hematology and Rheumatology, The Affiliated Hospital of Putian University, Fujian Province, China; School of Basic Medicine, Putian University, Fujian Province, China
| | - Yiyang Weng
- Pharmaceutical and Medical Technology College, Putian University, Fujian Province, China
| | - Haibin Wang
- Pharmaceutical and Medical Technology College, Putian University, Fujian Province, China
| | - Siting Zhan
- School of Basic Medicine, Putian University, Fujian Province, China
| | - Chaoqi Li
- Pharmaceutical and Medical Technology College, Putian University, Fujian Province, China
| | - Donghui Zheng
- Medical Image Center, The Affiliated Hospital of Putian University, Fujian Province, China
| | - Qi Lin
- Department of Pharmacy, The Affiliated Hospital of Putian University, Fujian Province, China.
| |
Collapse
|
18
|
Huang L. The role of IL-17 family cytokines in cardiac fibrosis. Front Cardiovasc Med 2024; 11:1470362. [PMID: 39502194 PMCID: PMC11534612 DOI: 10.3389/fcvm.2024.1470362] [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: 07/25/2024] [Accepted: 09/29/2024] [Indexed: 11/08/2024] Open
Abstract
Myocardial fibrosis is a common pathological feature in various cardiovascular diseases including myocardial infarction, heart failure, and myocarditis. Generally, persistent myocardial fibrosis correlates with poor prognosis and ranks among the leading causes of death globally. Currently, there is no effective treatment for myocardial fibrosis, partly due to its unclear pathogenic mechanism. Increasing studies have shown IL-17 family cytokines are strongly associated with the initiation and propagation of myocardial fibrosis. This review summarizes the expression, action, and signal transduction mechanisms of IL-17, focusing on its role in fibrosis associated with cardiovascular diseases such as myocardial infarction, heart failure, hypertension, diabetes, and myocarditis. It also discusses its potential as a therapeutic target, offering new insights for the clinical treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Liqing Huang
- Three Gorges University Hospital of Traditional Chinese Medicine & Yichang Hospital of Traditional Chinese Medicine, Yichang, China
| |
Collapse
|
19
|
Bédard-Matteau J, Soulé A, Liu KY, Fourcade L, Fraser DD, Emad A, Rousseau S. Circulating IL-17F, but not IL-17A, is elevated in severe COVID-19 and leads to an ERK1/2 and p38 MAPK-dependent increase in ICAM-1 cell surface expression and neutrophil adhesion on endothelial cells. Front Immunol 2024; 15:1452788. [PMID: 39493750 PMCID: PMC11527637 DOI: 10.3389/fimmu.2024.1452788] [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: 06/21/2024] [Accepted: 09/17/2024] [Indexed: 11/05/2024] Open
Abstract
Background Severe COVID-19 is associated with neutrophilic inflammation and immunothrombosis. Several members of the IL-17 cytokine family have been associated with neutrophilic inflammation and activation of the endothelium. Therefore, we investigated whether these cytokines were associated with COVID-19. Methods We investigated the association between COVID-19 and circulating plasma levels of IL-17 cytokine family members in participants to the Biobanque québécoise de la COVID-19 (BQC19), a prospective observational cohort and an independent cohort from Western University (London, Ontario). We measured the in vitro impact of IL-17F on intercellular adhesion molecule 1 (ICAM-1) cell surface expression and neutrophil adhesion on endothelial cells in culture. The contribution of two Mitogen Activated Protein Kinase (MAPK) pathways was determined using small molecule inhibitors PD184352 (a MKK1/MKK2 inhibitor) and BIRB0796 (a p38 MAPK inhibitor). Results We found increased IL-17D and IL-17F plasma levels when comparing SARS-CoV-2-positive vs negative hospitalized participants. Moreover, increased plasma levels of IL-17D, IL-17E and IL-17F were noted when comparing severe versus mild COVID-19. IL-17F, but not IL-17A, was significantly elevated in people with COVID-19 compared to healthy controls and with more severe disease. In vitro work on endothelial cells treated with IL-17F for 24h showed an increase cell surface expression of ICAM-1 accompanied by neutrophil adhesion. The introduction of two MAPK inhibitors significantly reduced the binding of neutrophils while also reducing ICAM-1 expression at the surface level of endothelial cells, but not its intracellular expression. Discussion Overall, these results have identified an association between two cytokines of the IL-17 family (IL-17D and IL-17F) with COVID-19 and disease severity. Considering that IL-17F stimulation promotes neutrophil adhesion to the endothelium in a MAPK-dependent manner, it is attractive to speculate that this pathway may contribute to pathogenic immunothrombosis in concert with other molecular effectors.
Collapse
Affiliation(s)
- Jérôme Bédard-Matteau
- The Meakins-Christie Laboratories, Research Institute of the McGill University Heath Centre, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Antoine Soulé
- Department of Electrical and Computer Engineering, McGill University, Montréal, QC, Canada
| | - Katelyn Yixiu Liu
- The Meakins-Christie Laboratories, Research Institute of the McGill University Heath Centre, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Lyvia Fourcade
- The Meakins-Christie Laboratories, Research Institute of the McGill University Heath Centre, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Douglas D. Fraser
- Children’s Health Research Institute & Lawson Health Research Institute, London, ON, Canada
- Department of Pediatrics, Western University, London, ON, Canada
| | - Amin Emad
- Department of Electrical and Computer Engineering, McGill University, Montréal, QC, Canada
- Mila, Quebec AI Institute, Montréal, QC, Canada
| | - Simon Rousseau
- The Meakins-Christie Laboratories, Research Institute of the McGill University Heath Centre, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| |
Collapse
|
20
|
Kulig P, Łuczkowska K, Machaliński B, Baumert B. Deep hematologic response to RD treatment in patients with multiple myeloma is associated with overexpression of IL-17R in CD138+ plasma cells. Sci Rep 2024; 14:23559. [PMID: 39384864 PMCID: PMC11464892 DOI: 10.1038/s41598-024-74558-3] [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: 04/09/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024] Open
Abstract
Lenalidomide (LEN) is widely used immunomodulatory drug (IMiD). Nonetheless, despite its efficacy, over time patients become resistant to LEN and relapse. Due to high clinical relevance, drug resistance in MM is being thoroughly investigated. However, less is known about predictors of good response to LEN-based treatment. The aim of this study was to identify molecular pathways associated with good and long response to LEN. The study included newly diagnosed MM patients (NDMM) and MM patients treated with first-line LEN and dexamethasone (RD) who achieved and least very good partial remission (VGPR). RNA was isolated from MM cells and new-generation sequencing was performed. Obtained results were validated with qRT-PCR. A global increase in gene expression was found in the RD group compared to NDMM, suggesting the involvement of epigenetic mechanisms. Moreover, upregulation of genes controlling the interaction within MM niche was detected. Next, genes controlling immune response were upregulated. In particular, the gene encoding the IL-17 receptor was overexpressed in the RD group which is a novel finding. This should be emphasized because IL-17-related signaling can potentially be targeted, providing the rationale for future research. Establishing the molecular background associated with long-lasting and profound response to LEN may improve LEN-based chemotherapy regimens and facilitate the development of adjuvant therapies to enhance its anti-MM activity.
Collapse
Affiliation(s)
- Piotr Kulig
- Department of General Pathology, Pomeranian Medical University, 70-111, Szczecin, Poland
- Pharmaceutical Facility of Pomeranian Medical University, 71-899, Szczecin, Poland
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University, 70-111, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, 70-111, Szczecin, Poland.
- Department of Hematology and Transplantology, Pomeranian Medical University, 71-252, Szczecin, Poland.
| | - Bartłomiej Baumert
- Department of Hematology and Transplantology, Pomeranian Medical University, 71-252, Szczecin, Poland.
| |
Collapse
|
21
|
Patra AK, Nayak S, Moharana A, Ojha P, Das SK, Akhtar J, Giri B, Singh S. Preliminary Investigation and Therapeutic Efficacy Determination of a Novel Anti-IL-17A Antibody, Indikizumab. Biologics 2024; 18:257-271. [PMID: 39319321 PMCID: PMC11420332 DOI: 10.2147/btt.s477752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 08/31/2024] [Indexed: 09/26/2024]
Abstract
Purpose The study aimed to develop and characterize Indikizumab, a novel humanized anti-IL-17A monoclonal antibody (mAb), for potential therapeutic use in inflammatory indications such as psoriasis, psoriatic arthritis, rheumatoid arthritis, and ankylosing spondylitis. Methods The research involved the purification of IL-17 isoforms, epitope mapping, affinity ranking, and comparative binding assessment of anti-IL-17 antibodies. The study also included cell-based neutralization assays and in vivo studies using mouse models to evaluate the efficacy of Indikizumab. Results Indikizumab demonstrated a high binding affinity (KD=27.2 pM) and specificity for IL-17A, with comparable potency to Secukinumab. In cell-based neutralization assays, Indikizumab effectively neutralized the effects of IL-17A and demonstrated a statistically significant reduction in plasma KC (Keratinocyte) levels in a mouse model. In imiquimod-induced psoriasis mouse model, Indikizumab showed potential in reducing the psoriasis index. Conclusion Indikizumab represents a promising therapeutic option for inflammatory indications with its high binding affinity, specificity for IL-17A, and effectiveness in neutralizing IL-17A effects in vivo.
Collapse
Affiliation(s)
- Ashok Kumar Patra
- Protein Expression Lab, Imgenex India Pvt. Ltd. E-5, Bhubaneswar, Odisha, India
| | - Shreenath Nayak
- Protein Expression Lab, Imgenex India Pvt. Ltd. E-5, Bhubaneswar, Odisha, India
| | - Anandita Moharana
- Protein Expression Lab, Imgenex India Pvt. Ltd. E-5, Bhubaneswar, Odisha, India
| | - Purusottam Ojha
- Protein Expression Lab, Imgenex India Pvt. Ltd. E-5, Bhubaneswar, Odisha, India
| | - Sanjeet Kumar Das
- Protein Expression Lab, Imgenex India Pvt. Ltd. E-5, Bhubaneswar, Odisha, India
| | - Jabed Akhtar
- Protein Expression Lab, Imgenex India Pvt. Ltd. E-5, Bhubaneswar, Odisha, India
| | - Bishwaranjan Giri
- Protein Expression Lab, Imgenex India Pvt. Ltd. E-5, Bhubaneswar, Odisha, India
| | - Sujay Singh
- Protein Expression Lab, Imgenex India Pvt. Ltd. E-5, Bhubaneswar, Odisha, India
- Sujan Biologics, Inc., San Diego, California, USA
| |
Collapse
|
22
|
Hu S, Li X, Li G, Li Z, He F, Tian G, Zhao X, Liu R. New Species and Cytotoxicity Mechanism of Halohydroxybenzonitrile Disinfection Byproducts in Drinking Water. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:15816-15826. [PMID: 39166926 DOI: 10.1021/acs.est.4c06163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Recently, seven dihalohydroxybenzonitriles (diHHBNs) have been determined as concerning nitrogenous aromatic disinfection byproducts (DBPs) in drinking water. Herein, eight new monohalohydroxybenzonitriles (monoHHBNs), including 3-chloro-2-hydroxybenzonitrile, 5-chloro-2-hydroxybenzonitrile, 3-chloro-4-hydroxybenzonitrile, 3-bromo-2-hydroxybenzonitrile, 5-bromo-2-hydroxybenzonitrile, 3-bromo-4-hydroxybenzonitrile, 5-iodo-2-hydroxybenzonitrile, and 3-iodo-4-hydroxybenzonitrile, were detected and identified in drinking water for the first time. Thereafter, the relative concentration-cytotoxicity contribution of each HHBN was calculated based on the acquired occurrence level and cytotoxicity data in this study, the genome-scale cytotoxicity mechanism was explored, and a quantitative structure-activity relationship (QSAR) model was developed. Results indicated that new monoHHBNs were present in drinking water at concentrations of 0.04-1.83 ng/L and exhibited higher cytotoxicity than some other monohalogenated aromatic DBPs. Notably, monoHHBNs showed concentration-cytotoxicity contribution comparable to diHHBNs, which have been previously identified as potential toxicity drivers in drinking water. Transcriptomic analysis revealed immunotoxicity and genotoxicity as dominant cytotoxicity mechanisms for HHBNs in Chinese hamster ovary (CHO-K1) cells, with potential carcinogenic effects. The QSAR model suggested oxidative stress and cellular uptake efficiency as important factors for their cytotoxicity, highlighting the importance of potential iodinated HHBNs in drinking water, such as 3,5-diiodo-2-hydroxybenzonitrile, for future studies. These findings are meaningful for better understanding the health risk and toxicological significance of HHBNs in drinking water.
Collapse
Affiliation(s)
- Shaoyang Hu
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, Qingdao 266237, China
| | - Xiangxiang Li
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, Qingdao 266237, China
| | - Guangzhao Li
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, Qingdao 266237, China
| | - Zhigang Li
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Falin He
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, Qingdao 266237, China
| | - Guang Tian
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, Qingdao 266237, China
| | - Xingchen Zhao
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, Qingdao 266237, China
| | - Rutao Liu
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, Qingdao 266237, China
| |
Collapse
|
23
|
Chen W, Tan M, Zhang H, Gao T, Ren J, Cheng S, Chen J. Signaling molecules in the microenvironment of hepatocellular carcinoma. Funct Integr Genomics 2024; 24:146. [PMID: 39207523 DOI: 10.1007/s10142-024-01427-7] [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: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Hepatocellular carcinoma (HCC) is a major fatal cancer that is known for its high recurrence and metastasis. An increasing number of studies have shown that the tumor microenvironment is closely related to the metastasis and invasion of HCC. The HCC microenvironment is a complex integrated system composed of cellular components, the extracellular matrix (ECM), and signaling molecules such as chemokines, growth factors, and cytokines, which are generally regarded as crucial molecules that regulate a series of important processes, such as the migration and invasion of HCC cells. Considering the crucial role of signaling molecules, this review aims to elucidate the regulatory effects of chemokines, growth factors, and cytokines on HCC cells in their microenvironment to provide important references for clarifying the development of HCC and exploring effective therapeutic targets.
Collapse
Affiliation(s)
- Wanjin Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Ming Tan
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Hui Zhang
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Tingting Gao
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Jihua Ren
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Shengtao Cheng
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China.
| | - Juan Chen
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China.
- College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
24
|
Ghosh D, Singh P, Reddy A, Patra P. Recurrent subcutaneous abscesses and pneumonia in a toddler with a novel pathogenic variation in IL-17RA gene. BMJ Case Rep 2024; 17:e261026. [PMID: 39179263 DOI: 10.1136/bcr-2024-261026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024] Open
Abstract
A toddler presented with recurrent subcutaneous abscesses, otitis media and pneumonia, requiring frequent hospitalisations and intravenous antimicrobials. He also had oral thrush and difficulty in gaining weight; hence, an underlying inborn error of immunity (IEI) was strongly suspected. The complete haemogram showed leucocytosis with neutrophilic predominance. Both erythrocyte sedimentation rate and C reactive protein were elevated. Klebsiella pneumoniae was isolated from blood culture. The dihydrorhodamine-123 assay was negative, and the immunoglobulin profile showed an increased IgG level. Whole exome sequencing revealed a novel homozygous pathogenic variation in the IL-17RA gene (c.2563G>A, p. Asp855Asn). He showed remarkable improvement following intravenous colistin and fluconazole with complete resolution of abscesses. Thus, it is prudent to consider the possibility of IL-17RA deficiency in children with a history of recurrent abscesses, skin ulcerations and pneumonia after excluding the common groups of IEI.
Collapse
Affiliation(s)
- Dipyaman Ghosh
- Pediatrics, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Pallavi Singh
- Pediatrics, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Aravind Reddy
- Pediatrics, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Pratap Patra
- Pediatrics, All India Institute of Medical Sciences, Patna, Bihar, India
| |
Collapse
|
25
|
Bhowmick S, Gupta S, Mondal S, Mallick AI. Activation of Antiviral Host Responses against Avian Influenza Virus and Remodeling of Gut Microbiota by rLAB Vector Expressing rIL-17A in Chickens. ACS Infect Dis 2024; 10:3026-3041. [PMID: 38970488 DOI: 10.1021/acsinfecdis.4c00377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Low-pathogenic avian influenza virus (LPAIV) remains the most common subtype of type-A influenza virus that causes moderate to severe infection in poultry with significant zoonotic and pandemic potential. Due to high mutability, increasing drug resistance, and limited vaccine availability, the conventional means to prevent intra- or interspecies transmission of AIV is highly challenging. As an alternative to control AIV infections, cytokine-based approaches to augment antiviral host defense have gained significant attention. However, the selective application of cytokines is critical since unregulated expression of cytokines, particularly proinflammatory ones, can cause substantial tissue damage during acute phases of immune responses. Moreover, depending on the type of cytokine and its impact on intestinal microbiota, outcomes of cytokine-gut microflora interaction can have a critical effect on overall host defense against AIV infections. Our recent study demonstrated some prominent roles of chicken IL-17A (ChIL-17A) in regulating antiviral host responses against AIV infection, however, in an in vitro model. For more detailed insights into ChIL-17A function, in the present study, we investigated whether ChIL-17A-meditated elevated antiviral host responses can translate into effective immune protection against AIV infection in an in vivo system. Moreover, considering the role of gut health in fostering innate or local host responses, we further studied the contributory relationships between gut microbiota and host immunity against AIV infection in chickens. For this, we employed a recombinant lactic acid-producing bacterial (LAB) vector, Lactococcus lactis, expressing ChIL-17A and analyzed the in vivo functionality in chickens against an LPAIV (A/H9N2) infection. Our study delineates that mucosal delivery of rL. lactis expressing ChIL-17A triggers proinflammatory signaling cascades and can drive a positive shift in phylum Firmicutes, along with a marked decline in phylum Actinobacteriota and Proteobacteria, favoring effective antiviral host responses against AIV infection in chickens. We propose that ChIL-17A-mediated selective expansion of beneficial gut microbiota might form a healthy microbial community that augments the effective immune protection against AIV infections in chickens.
Collapse
Affiliation(s)
- Sucharita Bhowmick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Subhadeep Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Samiran Mondal
- Department of Veterinary Pathology, West Bengal University of Animal and Fishery Sciences, Kolkata 700037, West Bengal, India
| | - Amirul Islam Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| |
Collapse
|
26
|
Abdelnabi MN, Hassan GS, Shoukry NH. Role of the type 3 cytokines IL-17 and IL-22 in modulating metabolic dysfunction-associated steatotic liver disease. Front Immunol 2024; 15:1437046. [PMID: 39156888 PMCID: PMC11327067 DOI: 10.3389/fimmu.2024.1437046] [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: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.
Collapse
Affiliation(s)
- Mohamed N. Abdelnabi
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
27
|
Xie E, Shen X, Yeo YH, Xing Z, Ebinger JE, Duan Y, Zhang Y, Cheng S, Ji F, Deng J. Exploring the underlying molecular mechanisms of acute myocardial infarction after SARS-CoV-2 infection. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 44:100417. [PMID: 39045234 PMCID: PMC11263507 DOI: 10.1016/j.ahjo.2024.100417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/20/2024] [Indexed: 07/25/2024]
Abstract
An increase in acute myocardial infarction (AMI)-related deaths has been reported during the COVID-19 pandemic. Despite evidence suggesting the association between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and AMI, the underlying mechanisms remain unclear. Here, we integrated mRNA and microRNA expression profiles related to SARS-CoV-2 infection and AMI from public databases. We then performed transcriptomic analysis using bioinformatics and systems biology approaches to explore the potential molecular mechanisms of SARS-CoV-2 infection affects AMI. First, twenty-one common differentially expressed genes (DEGs) were identified from SARS-CoV-2 infection and AMI patients in endothelial cells datasets and then we performed functional analysis to predict the roles of these DEGs. The functional analysis emphasized that the endothelial cell response to cytokine stimulus due to excessive inflammation was essential in these two diseases. Importantly, the tumor necrosis factor and interleukin-17 signaling pathways appeared to be integral factors in this mechanism. Interestingly, most of these common genes were also upregulated in transcriptomic datasets of SARS-CoV-2-infected cardiomyocytes, suggesting that these genes may be shared in cardiac- and vascular-related injuries. We subsequently built a protein-protein interaction network and extracted hub genes and essential modules from this network. At the transcriptional and post-transcriptional levels, regulatory networks with common DEGs were also constructed, and some key regulator signatures were further identified and validated. In summary, our research revealed that a highly activated inflammatory response in patients with COVID-19 might be a crucial factor for susceptibility to AMI and we identified some candidate genes and regulators that could be used as biomarkers or potential therapeutic targets.
Collapse
Affiliation(s)
- Enrui Xie
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Infectious Disease, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaotao Shen
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore
- Department of Genetics, Stanford University, California, USA
| | - Yee Hui Yeo
- Department of Genetics, Stanford University, California, USA
| | - Zixuan Xing
- Department of Infectious Disease, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Joseph E. Ebinger
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Yixuan Duan
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yue Zhang
- Department of Infectious Disease, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- The Eighth Hospital of Xi'an City, Xi'an Jiaotong University, Xi'an, China
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Fanpu Ji
- Department of Infectious Disease, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Medical Research Center of Infectious Diseases, Xi'an, China
| | - Jie Deng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
28
|
Lopalco G, Cito A, Venerito V, Iannone F, Proft F. The management of axial spondyloarthritis with cutting-edge therapies: advancements and innovations. Expert Opin Biol Ther 2024; 24:835-853. [PMID: 39109494 DOI: 10.1080/14712598.2024.2389987] [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: 05/28/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
INTRODUCTION Axial involvement in spondyloarthritis has significantly evolved from the original 1984 New York criteria for ankylosing spondylitis, leading to an improved understanding of axial spondyloarthritis (axSpA) as a disease continuum encompassing non- radiographic axSpA (nr-axSpA) and radiographic axSpA (r-axSpA). A clear definition for early axSpA has been established, underscoring the need for early intervention with biological and targeted synthetic drugs to mitigate pain, reduce functional impairment, and prevent radiographic progression. AREAS COVERED This review explores therapeutic strategies in axSpA management, focusing on biological and targeted synthetic therapies and recent advancements. Biologics targeting TNFα or IL-17 and targeted synthetic disease-modifying antirheumatic drugs (DMARDs) are primary treatment options. These therapies significantly impact clinical outcomes, radiographic progression, and patient-reported functional improvement. EXPERT OPINION AxSpA treatment has evolved significantly, offering various therapeutic options. Biological DMARDs, particularly TNFα inhibitors, have transformed treatment, significantly enhancing patient outcomes. However, challenges persist for patients unresponsive or intolerant to existing therapies. Emerging therapeutic targets promise to address these challenges. Comprehensive management strategies and personalized approaches, considering extra-articular manifestations and individual patient factors, are crucial for achieving optimal outcomes in axSpA management.
Collapse
Affiliation(s)
- Giuseppe Lopalco
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Andrea Cito
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Venerito
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Florenzo Iannone
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
29
|
Irawan A, Bionaz M. Liver Transcriptomic Profiles of Ruminant Species Fed Spent Hemp Biomass Containing Cannabinoids. Genes (Basel) 2024; 15:963. [PMID: 39062742 PMCID: PMC11275923 DOI: 10.3390/genes15070963] [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: 06/29/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
The inclusion of spent hemp biomass (SHB), an extracted byproduct from industrial cannabidiol (CBD) production, in the diets of dairy cows and lambs appears to be safe with minor effects on the metabolism, including a decrease in circulating cholesterol and increase bilirubinemia, both associated with liver metabolism. Those effects could be consequence of the presence of cannabinoids, particularly Δ9-tetrahydrocannabinol (THC) and CBD in the SHB. This study aimed to study the transcriptional profile of the liver of dairy cows and lambs fed SHB. Dairy cows received SHB or alfalfa pellet for four weeks of intervention (IP) and four weeks of withdrawal periods (WP). Finishing lambs were fed a control diet (CON), 10% (LH2), or 20% (HH2) SHB for 2 months or 1 month followed by 1-month SHB withdrawal (LH1 and HH1, respectively). RNA sequencing was performed, and the mRNA was annotated using the latest reference genomes. The RNAseq data were filtered, normalized for library size and composition, and statistically analyzed by DESeq2. The bioinformatic analysis was performed by using DAVID, Gene Set Enrichment Analysis (GSEA), and the Dynamic Impact Approach. Using a 0.2 FDR cut-off, we identified only ≤24 differentially expressed genes (DEG) in the liver by feeding SHB in dairy cows and a larger number of DEGs in lambs (from 71 in HH1 vs. CON to 552 in LH1 vs. CON). The KEGG analysis demonstrated that feeding SHB in dairy cows and lambs had relatively minor to moderate metabolic alterations in dairy cows and lambs mainly associated with amino acids and lipid metabolism whereas cholesterol synthesis was overall activated in lambs. GSEA identified activation of the PPAR signaling pathway only in dairy cows. We found an opposite effect on activation of metabolism of drug and xenobiotics by cytochrome P450 enzymes in dairy cows and lambs receiving less SHB but an inhibition in HH2 lambs. Immune system-related pathways were inhibited by feeding SHB in lambs, but the impact was minor. Cumulatively, inclusion of SHB containing cannabinoids in dairy and lambs demonstrate very little effects on the alteration of transcriptomic profile of the liver.
Collapse
Affiliation(s)
- Agung Irawan
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA;
- Animal Science Study Program, Universitas Sebelas Maret, Surakarta 57126, Indonesia
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA;
| |
Collapse
|
30
|
Hong L, Herjan T, Chen X, Zagore LL, Bulek K, Wang H, Yang CFJ, Licatalosi DD, Li X, Li X. Act1 drives chemoresistance via regulation of antioxidant RNA metabolism and redox homeostasis. J Exp Med 2024; 221:e20231442. [PMID: 38861022 PMCID: PMC11167376 DOI: 10.1084/jem.20231442] [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: 08/14/2023] [Revised: 02/20/2024] [Accepted: 05/09/2024] [Indexed: 06/12/2024] Open
Abstract
The IL-17 receptor adaptor molecule Act1, an RNA-binding protein, plays a critical role in IL-17-mediated cancer progression. Here, we report a novel mechanism of how IL-17/Act1 induces chemoresistance by modulating redox homeostasis through epitranscriptomic regulation of antioxidant RNA metabolism. Transcriptome-wide mapping of direct Act1-RNA interactions revealed that Act1 binds to the 5'UTR of antioxidant mRNAs and Wilms' tumor 1-associating protein (WTAP), a key regulator in m6A methyltransferase complex. Strikingly, Act1's binding sites are located in proximity to m6A modification sites, which allows Act1 to promote the recruitment of elF3G for cap-independent translation. Loss of Act1's RNA binding activity or Wtap knockdown abolished IL-17-induced m6A modification and translation of Wtap and antioxidant mRNAs, indicating a feedforward mechanism of the Act1-WTAP loop. We then developed antisense oligonucleotides (Wtap ASO) that specifically disrupt Act1's binding to Wtap mRNA, abolishing IL-17/Act1-WTAP-mediated antioxidant protein production during chemotherapy. Wtap ASO substantially increased the antitumor efficacy of cisplatin, demonstrating a potential therapeutic strategy for chemoresistance.
Collapse
Affiliation(s)
- Lingzi Hong
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tomasz Herjan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xing Chen
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Leah L. Zagore
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Katarzyna Bulek
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Han Wang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | - Donny D. Licatalosi
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Takeda Pharmaceutical Company, San Diego, CA, USA
| | - Xiaoxia Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xiao Li
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
31
|
Garcia Moreno AS, Guicciardi ME, Wixom AQ, Jessen E, Yang J, Ilyas SI, Bianchi JK, Pinto e Vairo F, Lazaridis KN, Gores GJ. IL-17 signaling in primary sclerosing cholangitis patient-derived organoids. Hepatol Commun 2024; 8:e0454. [PMID: 38829197 PMCID: PMC11150034 DOI: 10.1097/hc9.0000000000000454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/15/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND The pathogenesis of primary sclerosing cholangitis (PSC) is unclear, although studies implicate IL-17A as an inflammatory mediator in this disease. However, a direct assessment of IL-17 signaling in PSC cholangiocytes is lacking. In this study, we aimed to investigate and characterize the response of PSC extrahepatic cholangiocyte organoids (ECO) to IL-17A stimulation. METHODS Cholangiocytes obtained from patients with PSC and without PSC by endoscopic retrograde cholangiography were cultured as ECO. The ECO were treated with vehicle or IL-17A and assessed by transcriptomics, secretome analysis, and genome sequencing. RESULTS Unsupervised clustering of all integrated single-cell RNA sequencing data identified 8 cholangiocyte clusters that did not differ between PSC and non-PSC ECO. However, PSC ECO cells demonstrated a robust response to IL-17 treatment, as noted by an increased number of differentially expressed genes by transcriptomics and more abundant chemokine and cytokine expression and secretion. After rigorous filtering, genome sequencing identified candidate somatic variants shared among PSC ECO from unrelated individuals. However, no candidate rare variants in genes regulating the IL-17 pathway were identified, but rare variants regulating the MAPK signaling pathway were present in all PSC ECO. CONCLUSIONS PSC and non-PSC patient-derived ECO respond differently to IL-17 stimulation, implicating this pathway in the pathogenesis of PSC.
Collapse
Affiliation(s)
- Ana S. Garcia Moreno
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Maria E. Guicciardi
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander Q. Wixom
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Erik Jessen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Jingchun Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Sumera I. Ilyas
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jackie K. Bianchi
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Filippo Pinto e Vairo
- Center for Individualized Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| | - Konstantinos N. Lazaridis
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
32
|
Pasamba EC, Orda MA, Villanueva BHA, Tsai PW, Tayo LL. Transcriptomic Analysis of Hub Genes Reveals Associated Inflammatory Pathways in Estrogen-Dependent Gynecological Diseases. BIOLOGY 2024; 13:397. [PMID: 38927277 PMCID: PMC11201105 DOI: 10.3390/biology13060397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024]
Abstract
Gynecological diseases are triggered by aberrant molecular pathways that alter gene expression, hormonal balance, and cellular signaling pathways, which may lead to long-term physiological consequences. This study was able to identify highly preserved modules and key hub genes that are mainly associated with gynecological diseases, represented by endometriosis (EM), ovarian cancer (OC), cervical cancer (CC), and endometrial cancer (EC), through the weighted gene co-expression network analysis (WGCNA) of microarray datasets sourced from the Gene Expression Omnibus (GEO) database. Five highly preserved modules were observed across the EM (GSE51981), OC (GSE63885), CC (GSE63514), and EC (GSE17025) datasets. The functional annotation and pathway enrichment analysis revealed that the highly preserved modules were heavily involved in several inflammatory pathways that are associated with transcription dysregulation, such as NF-kB signaling, JAK-STAT signaling, MAPK-ERK signaling, and mTOR signaling pathways. Furthermore, the results also include pathways that are relevant in gynecological disease prognosis through viral infections. Mutations in the ESR1 gene that encodes for ERα, which were shown to also affect signaling pathways involved in inflammation, further indicate its importance in gynecological disease prognosis. Potential drugs were screened through the Drug Repurposing Encyclopedia (DRE) based on the up-and downregulated hub genes, wherein a bacterial ribosomal subunit inhibitor and a benzodiazepine receptor agonist were the top candidates. Other drug candidates include a dihydrofolate reductase inhibitor, glucocorticoid receptor agonists, cholinergic receptor agonists, selective serotonin reuptake inhibitors, sterol demethylase inhibitors, a bacterial antifolate, and serotonin receptor antagonist drugs which have known anti-inflammatory effects, demonstrating that the gene network highlights specific inflammatory pathways as a therapeutic avenue in designing drug candidates for gynecological diseases.
Collapse
Affiliation(s)
- Elaine C. Pasamba
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines; (E.C.P.); (M.A.O.); (B.H.A.V.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
| | - Marco A. Orda
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines; (E.C.P.); (M.A.O.); (B.H.A.V.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
| | - Brian Harvey Avanceña Villanueva
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines; (E.C.P.); (M.A.O.); (B.H.A.V.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
| | - Po-Wei Tsai
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan;
| | - Lemmuel L. Tayo
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines; (E.C.P.); (M.A.O.); (B.H.A.V.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
- Department of Biology, School of Health Sciences, Mapúa University, Makati City 1203, Philippines
| |
Collapse
|
33
|
Dascălu RC, Bărbulescu AL, Stoica LE, Dinescu ȘC, Biță CE, Popoviciu HV, Ionescu RA, Vreju FA. Review: A Contemporary, Multifaced Insight into Psoriasis Pathogenesis. J Pers Med 2024; 14:535. [PMID: 38793117 PMCID: PMC11122105 DOI: 10.3390/jpm14050535] [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: 03/26/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Psoriasis is a chronic recurrent inflammatory autoimmune pathology with a significant genetic component and several interferences of immunological cells and their cytokines. The complex orchestration of psoriasis pathogenesis is related to the synergic effect of immune cells, polygenic alterations, autoantigens, and several other external factors. The major act of the IL-23/IL-17 axis, strongly influencing the inflammatory pattern established during the disease activity, is visible as a continuous perpetuation of the pro-inflammatory response and keratinocyte activation and proliferation, leading to the development of psoriatic lesions. Genome-wide association studies (GWASs) offer a better view of psoriasis pathogenic pathways, with approximately one-third of psoriasis's genetic impact on psoriasis development associated with the MHC region, with genetic loci located on chromosome 6. The most eloquent genetic factor of psoriasis, PSORS1, was identified in the MHC I site. Among the several factors involved in its complex etiology, dysbiosis, due to genetic or external stimulus, induces a burst of pro-inflammatory consequences; both the cutaneous and gut microbiome get involved in the psoriasis pathogenic process. Cutting-edge research studies and comprehensive insights into psoriasis pathogenesis, fostering novel genetic, epigenetic, and immunological factors, have generated a spectacular improvement over the past decades, securing the path toward a specific and targeted immunotherapeutic approach and delayed progression to inflammatory arthritis. This review aimed to offer insight into various domains that underline the pathogenesis of psoriasis and how they influence disease development and evolution. The pathogenesis mechanism of psoriasis is multifaceted and involves an interplay of cellular and humoral immunity, which affects susceptible microbiota and the genetic background. An in-depth understanding of the role of pathogenic factors forms the basis for developing novel and individualized therapeutic targets that can improve disease management.
Collapse
Affiliation(s)
- Rucsandra Cristina Dascălu
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.C.D.); (C.E.B.); (F.A.V.)
| | - Andreea Lili Bărbulescu
- Department of Pharmacology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Loredana Elena Stoica
- Department of Dermatology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Ștefan Cristian Dinescu
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.C.D.); (C.E.B.); (F.A.V.)
| | - Cristina Elena Biță
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.C.D.); (C.E.B.); (F.A.V.)
| | - Horațiu Valeriu Popoviciu
- Department of Rheumatology, BFK and Medical Rehabilitation, University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Mures, Romania;
| | - Răzvan Adrian Ionescu
- Third Internal Medicine Department, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Florentin Ananu Vreju
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.C.D.); (C.E.B.); (F.A.V.)
| |
Collapse
|
34
|
Roy S, Saha P, Bose D, Trivedi A, More M, Lin C, Wu J, Oakes M, Chatterjee S. Periodic heat waves-induced neuronal etiology in the elderly is mediated by gut-liver-brain axis: a transcriptome profiling approach. Sci Rep 2024; 14:10555. [PMID: 38719902 PMCID: PMC11079080 DOI: 10.1038/s41598-024-60664-9] [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: 01/12/2024] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
Heat stress exposure in intermittent heat waves and subsequent exposure during war theaters pose a clinical challenge that can lead to multi-organ dysfunction and long-term complications in the elderly. Using an aged mouse model and high-throughput sequencing, this study investigated the molecular dynamics of the liver-brain connection during heat stress exposure. Distinctive gene expression patterns induced by periodic heat stress emerged in both brain and liver tissues. An altered transcriptome profile showed heat stress-induced altered acute phase response pathways, causing neural, hepatic, and systemic inflammation and impaired synaptic plasticity. Results also demonstrated that proinflammatory molecules such as S100B, IL-17, IL-33, and neurological disease signaling pathways were upregulated, while protective pathways like aryl hydrocarbon receptor signaling were downregulated. In parallel, Rantes, IRF7, NOD1/2, TREM1, and hepatic injury signaling pathways were upregulated. Furthermore, current research identified Orosomucoid 2 (ORM2) in the liver as one of the mediators of the liver-brain axis due to heat exposure. In conclusion, the transcriptome profiling in elderly heat-stressed mice revealed a coordinated network of liver-brain axis pathways with increased hepatic ORM2 secretion, possibly due to gut inflammation and dysbiosis. The above secretion of ORM2 may impact the brain through a leaky blood-brain barrier, thus emphasizing intricate multi-organ crosstalk.
Collapse
Affiliation(s)
- Subhajit Roy
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA, 92697, USA
| | - Punnag Saha
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA, 92697, USA
| | - Dipro Bose
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA, 92697, USA
| | - Ayushi Trivedi
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA, 92697, USA
| | - Madhura More
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA, 92697, USA
| | - Christina Lin
- Genomics Research and Technology Hub, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Jie Wu
- Genomics Research and Technology Hub, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Melanie Oakes
- Genomics Research and Technology Hub, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA, 92697, USA.
- Division of Infectious Diseases, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Long Beach VA Medical Center, Long Beach, CA, 90822, USA.
| |
Collapse
|
35
|
Chen S, Fan H, Ran C, Hong Y, Feng H, Yue Z, Zhang H, Pontarotti P, Xu A, Huang S. The IL-17 pathway intertwines with neurotrophin and TLR/IL-1R pathways since its domain shuffling origin. Proc Natl Acad Sci U S A 2024; 121:e2400903121. [PMID: 38683992 PMCID: PMC11087794 DOI: 10.1073/pnas.2400903121] [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: 02/02/2024] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
The IL-17 pathway displays remarkably diverse functional modes between different subphyla, classes, and even orders, yet its driving factors remains elusive. Here, we demonstrate that the IL-17 pathway originated through domain shuffling between a Toll-like receptor (TLR)/IL-1R pathway and a neurotrophin-RTK (receptor-tyrosine-kinase) pathway (a Trunk-Torso pathway). Unlike other new pathways that evolve independently, the IL-17 pathway remains intertwined with its donor pathways throughout later evolution. This intertwining not only influenced the gains and losses of domains and components in the pathway but also drove the diversification of the pathway's functional modes among animal lineages. For instance, we reveal that the crustacean female sex hormone, a neurotrophin inducing sex differentiation, could interact with IL-17Rs and thus be classified as true IL-17s. Additionally, the insect prothoracicotropic hormone, a neurotrophin initiating ecdysis in Drosophila by binding to Torso, could bind to IL-17Rs in other insects. Furthermore, IL-17R and TLR/IL-1R pathways maintain crosstalk in amphioxus and zebrafish. Moreover, the loss of the Death domain in the pathway adaptor connection to IκB kinase and stress-activated protein kinase (CIKSs) dramatically reduced their abilities to activate nuclear factor-kappaB (NF-κB) and activator protein 1 (AP-1) in amphioxus and zebrafish. Reinstating this Death domain not only enhanced NF-κB/AP-1 activation but also strengthened anti-bacterial immunity in zebrafish larvae. This could explain why the mammalian IL-17 pathway, whose CIKS also lacks Death, is considered a weak signaling activator, relying on synergies with other pathways. Our findings provide insights into the functional diversity of the IL-17 pathway and unveil evolutionary principles that could govern the pathway and be used to redesign and manipulate it.
Collapse
Affiliation(s)
- Shenghui Chen
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao266237, China
| | - Huiping Fan
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Chenrui Ran
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Yun Hong
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Huixiong Feng
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Zirui Yue
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Hao Zhang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Pierre Pontarotti
- MEPHI (Microbes, Evolution, Phylogénie et Infection), Aix Marseille Université, Marseille, France
| | - Anlong Xu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing100029, China
| | - Shengfeng Huang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao266237, China
| |
Collapse
|
36
|
Jin B, Moududee SA, Ge D, Zhou P, Wang AR, Liu YZ, You Z. SCF FBXW11 Complex Targets Interleukin-17 Receptor A for Ubiquitin-Proteasome-Mediated Degradation. Biomedicines 2024; 12:755. [PMID: 38672111 PMCID: PMC11047997 DOI: 10.3390/biomedicines12040755] [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: 02/28/2024] [Revised: 03/23/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Interleukin-17 (IL-17) is a pro-inflammatory cytokine that participates in innate and adaptive immune responses and plays an important role in host defense, autoimmune diseases, tissue regeneration, metabolic regulation, and tumor progression. Post-translational modifications (PTMs) are crucial for protein function, stability, cellular localization, cellular transduction, and cell death. However, PTMs of IL-17 receptor A (IL-17RA) have not been investigated. Here, we show that human IL-17RA was targeted by F-box and WD repeat domain-containing 11 (FBXW11) for ubiquitination, followed by proteasome-mediated degradation. We used bioinformatics tools and biochemical techniques to determine that FBXW11 ubiquitinated IL-17RA through a lysine 27-linked polyubiquitin chain, targeting IL-17RA for proteasomal degradation. Domain 665-804 of IL-17RA was critical for interaction with FBXW11 and subsequent ubiquitination. Our study demonstrates that FBXW11 regulates IL-17 signaling pathways at the IL-17RA level.
Collapse
Affiliation(s)
- Ben Jin
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70112, USA; (B.J.); (S.A.M.)
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA 70112, USA
| | - Sayed Ala Moududee
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70112, USA; (B.J.); (S.A.M.)
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA 70112, USA
| | - Dongxia Ge
- Department of Orthopaedic Surgery, Tulane University, New Orleans, LA 70112, USA;
| | - Pengbo Zhou
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Alun R. Wang
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Yao-Zhong Liu
- Department of Biostatistics and Data Science, Tulane University, New Orleans, LA 70112, USA;
| | - Zongbing You
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70112, USA; (B.J.); (S.A.M.)
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA 70112, USA
- Department of Orthopaedic Surgery, Tulane University, New Orleans, LA 70112, USA;
- Tulane Cancer Center and Louisiana Cancer Research Consortium, Tulane University, New Orleans, LA 70112, USA
- Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, LA 70112, USA
- Tulane Center for Aging, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
37
|
Amormino C, Russo E, Tedeschi V, Fiorillo MT, Paiardini A, Spallotta F, Rosanò L, Tuosto L, Kunkl M. Targeting staphylococcal enterotoxin B binding to CD28 as a new strategy for dampening superantigen-mediated intestinal epithelial barrier dysfunctions. Front Immunol 2024; 15:1365074. [PMID: 38510259 PMCID: PMC10951378 DOI: 10.3389/fimmu.2024.1365074] [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: 01/03/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Staphylococcus aureus is a gram-positive bacterium that may cause intestinal inflammation by secreting enterotoxins, which commonly cause food-poisoning and gastrointestinal injuries. Staphylococcal enterotoxin B (SEB) acts as a superantigen (SAg) by binding in a bivalent manner the T-cell receptor (TCR) and the costimulatory receptor CD28, thus stimulating T cells to produce large amounts of inflammatory cytokines, which may affect intestinal epithelial barrier integrity and functions. However, the role of T cell-mediated SEB inflammatory activity remains unknown. Here we show that inflammatory cytokines produced by T cells following SEB stimulation induce dysfunctions in Caco-2 intestinal epithelial cells by promoting actin cytoskeleton remodelling and epithelial cell-cell junction down-regulation. We also found that SEB-activated inflammatory T cells promote the up-regulation of epithelial-mesenchymal transition transcription factors (EMT-TFs) in a nuclear factor-κB (NF-κB)- and STAT3-dependent manner. Finally, by using a structure-based design approach, we identified a SEB mimetic peptide (pSEB116-132) that, by blocking the binding of SEB to CD28, dampens inflammatory-mediated dysregulation of intestinal epithelial barrier.
Collapse
Affiliation(s)
- Carola Amormino
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Emanuela Russo
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Valentina Tedeschi
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Alessandro Paiardini
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, Rome, Italy
| | - Francesco Spallotta
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Laboratory affiliated to Instituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Laura Rosanò
- Institute of Molecular Biology and Pathology, CNR, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Martina Kunkl
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
38
|
Zhang T, Huo H, Zhang Y, Tao J, Yang J, Rong X, Yang Y. Th17 cells: A new target in kidney disease research. Int Rev Immunol 2024; 43:263-279. [PMID: 38439681 DOI: 10.1080/08830185.2024.2321901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 03/06/2024]
Abstract
Type 17 T helper (Th17) cells, which are a subtype of CD4+ T helper cells, secrete pro-inflammatory cytokines such as IL-17A, IL-17F, IL-21, IL-22, and GM-CSF, which play crucial roles in immune defence and protection against fungal and extracellular pathogen invasion. However, dysfunction of Th17 cell immunity mediates inflammatory responses and exacerbates tissue damage. This pathological process initiated by Th17 cells is common in kidney diseases associated with renal injury, such as glomerulonephritis, lupus nephritis, IgA nephropathy, hypertensive nephropathy, diabetic kidney disease and acute kidney injury. Therefore, targeting Th17 cells to treat kidney diseases has been a hot topic in recent years. This article reviews the mechanisms of Th17 cell-mediated inflammation and autoimmune responses in kidney diseases and discusses the related clinical drugs that modulate Th17 cell fate in kidney disease treatment.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hongyan Huo
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yinghui Zhang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie Tao
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Junzheng Yang
- Guangdong Nephrotic Drug Engineering Technology Research Center, The R&D Center of Drug for Renal Diseases, Consun Pharmaceutical Group, Guangzhou, Guangdong, China
| | - Xianglu Rong
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yiqi Yang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
39
|
Hou CY, Hsieh CC, Hung YC, Hsu CC, Hsieh CW, Yu SH, Cheng KC. Evaluation of the amelioration effect of Ganoderma formosanum extract on delaying PM2.5 damage to lung macrophages. Mol Nutr Food Res 2024; 68:e2300667. [PMID: 38282089 DOI: 10.1002/mnfr.202300667] [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] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/26/2023] [Indexed: 01/30/2024]
Abstract
SCOPE Particulate matter (PM) contains toxic organic matter and heavy metals that enter the entire body through blood flow and may cause mortality. Ganoderma formosanum mycelium, a valuable traditional Chinese medicine that has been used since ancient times, contains various active ingredients that can effectively impede inflammatory responses on murine alveolar macrophages induced by PM particles. METHODS AND RESULTS An experimental study assessing the effect of G. formosanum mycelium extract's water fraction (WA) on PM-exposed murine alveolar macrophages using ROS measurement shows that WA reduces intracellular ROS by 12% and increases cell viability by 16% when induced by PM particles. According to RNA-Sequencing, western blotting, and real-time qPCR are conducted to analyze the metabolic pathway. The WA reduces the protein ratio in p-NF-κB/NF-κB by 18% and decreases the expression of inflammatory genes, including IL-1β by 38%, IL-6 by 29%, and TNF-α by 19%. Finally, the identification of seven types of anti-inflammatory compounds in the WA fraction is achieved through UHPLC-ESI-Orbitrap-Elite-MS/MS analysis. These compounds include anti-inflammatory compounds, namely thiamine, adenosine 5'-monophosphate, pipecolic acid, L-pyroglutamic acid, acetyl-L-carnitine, D-mannitol, and L-malic acid. CONCLUSIONS The study suggests that the WA has the potential to alleviate the PM -induced damage in alveolar macrophages, demonstrating its anti-inflammatory properties.
Collapse
Affiliation(s)
- Chih-Yao Hou
- Department of Seafood Science, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Chen-Che Hsieh
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan
| | - Yin-Ci Hung
- Institute of Food Science Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Chang-Wei Hsieh
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung, 40227, Taiwan
| | - Shu-Han Yu
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan
| | - Kuan-Chen Cheng
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan
- Institute of Food Science Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan
- Department of Optometry, Asia University, 500, Lioufeng Rd., Wufeng, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, 91, Hsueh-Shih Road, Taichung, Taiwan
| |
Collapse
|
40
|
Pantoja CJ, Li H, Rodante J, Keel A, Sorokin AV, Svedbom A, Teague HL, Stahle M, Mehta NN, Playford MP. Serum Beta-Defensin-2 is a biomarker for psoriasis but not subclinical atherosclerosis: Role of IL17a, PI-3 kinase and Rac1. JEADV CLINICAL PRACTICE 2024; 3:150-159. [PMID: 38646149 PMCID: PMC11031204 DOI: 10.1002/jvc2.278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/20/2023] [Indexed: 04/23/2024]
Abstract
Background Beta-defensins (BDs) are antimicrobial peptides secreted upon epithelial injury. Both chemotactic and antimicrobial properties of BDs function as initial steps in host defense and prime the adaptive immune system in the body. Psoriasis, a chronic immune-mediated inflammatory disease, has both visible cutaneous manifestations as well as known associations with higher incidence of cardiometabolic complications and vascular inflammation. Objectives We aimed to investigate the circulating expression of beta-defensin-2 (BD2) in psoriasis at baseline compared to control subjects, along with changes in BD2 levels following biologic treatment at one-year. The contribution of BD2 to subclinical atherosclerosis is also assessed. In addition, we have sought to unravel signaling mechanisms linking inflammation with BD2 expression. Methods Multimodality imaging as well inflammatory biomarker assays were performed in biologic naïve psoriasis (n=71) and non-psoriasis (n=53) subjects. A subset of psoriasis patients were followed for one-year after biological intervention (anti-Tumor Necrosis Factor-α (TNFα), n=30; anti-Interleukin17A (IL17A), n=21). Measurements of circulating BD2 were completed by Enzyme-Linked Immunosorbent Assay (ELISA). Using HaCaT transformed keratinocytes, expression of BD2 upon cytokine treatment was assessed by quantitative polymerase chain reaction (qPCR) and ELISA. Results Herein, we confirm that human circulating BD2 levels associate with psoriasis, which attenuate upon biologic interventions (anti-TNFα, anti-IL-17A). A link between circulating BD2 and sub-clinical atherosclerosis markers was not observed. Furthermore, we demonstrate that IL-17A-driven BD2 expression occurs in a Phosphatidylinositol 3-kinase (PI3-kinase) and Rac1 GTPase-dependent manner. Conclusions Our findings expand on the potential role of BD2 as a tractable biomarker in psoriasis patients and describes the role of an IL-17A-PI3-kinase/Rac signaling axis in regulating BD2 levels in keratinocytes.
Collapse
Affiliation(s)
- CJ. Pantoja
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - H. Li
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - J. Rodante
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - A. Keel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - AV. Sorokin
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - A. Svedbom
- Division of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - HL. Teague
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - M. Stahle
- Division of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - NN. Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - MP. Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| |
Collapse
|
41
|
Amira G, Akram D, Fadoua M, Bilel N, Alya B, Khalil BS, Monia SK, Fatma S, Habib HM, Nathalie B, Raja TM. Imbalance of TH17/TREG cells in Tunisian patients with systemic sclerosis. Presse Med 2024; 53:104221. [PMID: 38161053 DOI: 10.1016/j.lpm.2023.104221] [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: 03/18/2022] [Revised: 08/07/2022] [Accepted: 11/22/2022] [Indexed: 01/03/2024] Open
Abstract
Fibrosis is a pathological manifestation in which connective tissue replaces normal one. It can affect many tissues from the skin to internal organs such as the lungs. Manifestations of pulmonary involvement can be pulmonary arterial hypertension or pulmonary fibrosis. The latter one is currently the leading cause of death in various autoimmune diseases, including systemic sclerosis. Our study group consists of 50 patients with systemic sclerosis: 24 with limited cutaneous form and 26 with diffuse cutaneous form. This cohort was compared to 50 healthy controls (age and sex matched); our aim is to explore the distribution of TH17 cells (TH17) as well as regulatory T cells (TREG) and study their correlation with the disease's progress. Our results show an increase for IL17A in patients compared to controls and that this increase is correlated with a specific clinical involvement: Pulmonary fibrosis. This correlation suggests a crucial role of IL17A in fibrosis especially in systemic sclerosis. In addition, we have shown that the percentages of TH17 cells are higher in patients; however, the percentages of TREG cells are similar between patients and controls. A study of TREG cell activity showed that TREG lost suppressive activity by inactivating the FOXP3 transcription factor. This proves that despite their presence, TREG does not adequately carry out their regulatory activity. Finally, we analyzed the correlation between TH17/TREG and clinical damage; the results show a positive correlation with pulmonary involvement proving the role of TH17/TREG balance in induced fibrosis in systemic sclerosis. No significative difference was observed, for all the parameters, between the two different forms of the disease. In conclusion, the results associated with the TH17/TREG scale and their correlations with fibrosis in systemic sclerosis open a way for new tools to manage this autoimmune disease, which up to today has neither treatment nor accurate diagnosis.
Collapse
Affiliation(s)
- Gabsi Amira
- Laboratory of Genetics Immunology and Human Pathology, University of Tunis El Manar, LR05ES05, Tunis 2092, Tunisia; Aix Marseille University, INSERM, C2VN UMR1263, Marseille, France.
| | - Dlala Akram
- Laboratory of Genetics Immunology and Human Pathology, University of Tunis El Manar, LR05ES05, Tunis 2092, Tunisia
| | - Missaoui Fadoua
- Laboratory of Genetics Immunology and Human Pathology, University of Tunis El Manar, LR05ES05, Tunis 2092, Tunisia
| | - Neili Bilel
- Laboratory of Genetics Immunology and Human Pathology, University of Tunis El Manar, LR05ES05, Tunis 2092, Tunisia
| | - Boutaba Alya
- Laboratory of Genetics Immunology and Human Pathology, University of Tunis El Manar, LR05ES05, Tunis 2092, Tunisia
| | - Ben Salem Khalil
- Laboratory of Genetics Immunology and Human Pathology, University of Tunis El Manar, LR05ES05, Tunis 2092, Tunisia
| | - Smiti Khanfir Monia
- Internal medicine service, University hospital center LA RABTA, Tunis 1007, Tunisia; University of Tunis EL MANAR, Faculty of medicine Tunis, Tunis 1007, Tunisia
| | - Said Fatma
- Internal medicine service, University hospital center LA RABTA, Tunis 1007, Tunisia; University of Tunis EL MANAR, Faculty of medicine Tunis, Tunis 1007, Tunisia
| | - Houman Mohamed Habib
- Internal medicine service, University hospital center LA RABTA, Tunis 1007, Tunisia; University of Tunis EL MANAR, Faculty of medicine Tunis, Tunis 1007, Tunisia
| | - Bardin Nathalie
- Aix Marseille University, INSERM, C2VN UMR1263, Marseille, France; Laboratory of immunology, University hospital La Conception Marseille France, France
| | - Triki Marrakchi Raja
- Laboratory of Genetics Immunology and Human Pathology, University of Tunis El Manar, LR05ES05, Tunis 2092, Tunisia
| |
Collapse
|
42
|
Yang M, Zhang CY. Interleukins in liver disease treatment. World J Hepatol 2024; 16:140-145. [PMID: 38495285 PMCID: PMC10941743 DOI: 10.4254/wjh.v16.i2.140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 02/27/2024] Open
Abstract
Cytokines play pleiotropic roles in human health and disease by regulating both innate and adaptive immune responses. Interleukins (ILs), a large group of cytokines, can be divided into seven families, including IL-1, IL-2, IL-6, IL-8, IL-10, IL-12, and IL-17 families. Here, we review the functions of ILs in the pathogenesis and resolution of liver diseases, such as liver inflammation (e.g., IL-35), alcohol-related liver disease (e.g., IL-11), non-alcoholic steatohepatitis (e.g., IL-22), liver fibrosis (e.g., Il-17a), and liver cancer (e.g., IL-8). Overall, IL-1 family members are implicated in liver inflammation induced by different etiologies, such as alcohol consumption, high-fat diet, and hepatitis viruses. IL-2 family members mainly regulate T lymphocyte and NK cell proliferation and activation, and the differentiation of T cells. IL-6 family cytokines play important roles in acute phase response in liver infection, liver regeneration, and metabolic regulation, as well as lymphocyte activation. IL-8, also known as CXCL8, is activated in chronic liver diseases, which is associated with the accumulation of neutrophils and macrophages. IL-10 family members contribute key roles to liver immune tolerance and immunosuppression in liver disease. IL-12 family cytokines influence T-cell differentiation and play an essential role in autoimmune liver disease. IL-17 subfamilies contribute to infection defense, liver inflammation, and Th17 cell differentiation. ILs interact with different type I and type II cytokine receptors to regulate intracellular signaling pathways that mediate their functions. However, most clinical studies are only performed to evaluate IL-mediated therapies on alcohol and hepatitis virus infection-induced hepatitis. More pre-clinical and clinical studies are required to evaluate IL-mediated monotherapy and synergistic therapies.
Collapse
Affiliation(s)
- Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States.
| | - Chun-Ye Zhang
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, United States
| |
Collapse
|
43
|
Bai J, Tan BK, Kato A. Endotypic heterogeneity and pathogenesis in chronic rhinosinusitis. Curr Opin Allergy Clin Immunol 2024; 24:1-8. [PMID: 37966157 PMCID: PMC10873077 DOI: 10.1097/aci.0000000000000954] [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] [Indexed: 11/16/2023]
Abstract
PURPOSE OF REVIEW This review aims to provide updates in realms of endotypic heterogeneity, pathogenesis at the molecular level, potential of biomarkers, and cutting-edge scope of biologics in CRS. RECENT FINDINGS High-dimensional analyses, such as transcriptomes, and machine learning, have significantly enhanced CRS endotyping, uncovering diverse pathogenetic mechanisms contributing to its heterogeneity. The dynamic process of epithelial remodeling in CRS pathogenesis has gained more clarity and support as exemplified by IL-13 and oncostatin M (OSM) that are shown intricately linked to epithelial barrier dysfunction. Moreover, anti-dsDNA autoantibody, BAFF, periostin, and cystatin SN show promise as potentials biomarkers, offering diagnostic and prognostic value for CRS. SUMMARY The identification of inflammatory molecules involved in endotype specific signaling pathways provides insights into the underlying mechanisms and verifiable biomarkers for diagnosis and prediction of disease severity. More comprehensive clinical studies should be conducted to facilitate biologics from bench to bedside in treating CRS.
Collapse
Affiliation(s)
- Junqin Bai
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Bruce K. Tan
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Atsushi Kato
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
44
|
Pratelli G, Tamburini B, Badami GD, Lo Pizzo M, De Blasio A, Carlisi D, Di Liberto D. Cow's Milk: A Benefit for Human Health? Omics Tools and Precision Nutrition for Lactose Intolerance Management. Nutrients 2024; 16:320. [PMID: 38276558 PMCID: PMC10819418 DOI: 10.3390/nu16020320] [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: 12/28/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Cow's milk (CM) is a healthy food consumed worldwide by individuals of all ages. Unfortunately, "lactase-deficient" individuals cannot digest milk's main carbohydrate, lactose, depriving themselves of highly beneficial milk proteins like casein, lactoalbumin, and lactoglobulin due to lactose intolerance (LI), while other individuals develop allergies specifically against these proteins (CMPA). The management of these conditions differs, and an inappropriate diagnosis or treatment may have significant implications for the patients, especially if they are infants or very young children, resulting in unnecessary dietary restrictions or avoidable adverse reactions. Omics technologies play a pivotal role in elucidating the intricate interactions between nutrients and the human body, spanning from genetic factors to the microbiota profile and metabolites. This comprehensive approach enables the precise delineation and identification of distinct cohorts of individuals with specific dietary requirements, so that tailored nutrition strategies can be developed. This is what is called personalized nutrition or precision nutrition (PN), the area of nutrition that focuses on the effects of nutrients on the genome, proteome, and metabolome, promoting well-being and health, preventing diseases, reducing chronic disease incidence, and increasing life expectancy. Here, we report the opinion of the scientific community proposing to replace the "one size fits all" approach with tailor-made nutrition programs, designed by integrating nutrigenomic data together with clinical parameters and microbiota profiles, taking into account the individual lactose tolerance threshold and needs in terms of specific nutrients intake. This customized approach could help LI patients to improve their quality of life, overcoming depression or anxiety often resulting from the individual perception of this condition as different from a normal state.
Collapse
Affiliation(s)
- Giovanni Pratelli
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy; (G.P.); (D.C.)
| | - Bartolo Tamburini
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90127 Palermo, Italy;
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, 90127 Palermo, Italy; (G.D.B.); (M.L.P.)
| | - Giusto Davide Badami
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, 90127 Palermo, Italy; (G.D.B.); (M.L.P.)
| | - Marianna Lo Pizzo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, 90127 Palermo, Italy; (G.D.B.); (M.L.P.)
| | - Anna De Blasio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Laboratory of Biochemistry, University of Palermo, 90127 Palermo, Italy;
| | - Daniela Carlisi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy; (G.P.); (D.C.)
| | - Diana Di Liberto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy; (G.P.); (D.C.)
| |
Collapse
|
45
|
Sisto M, Lisi S. Targeting Interleukin-17 as a Novel Treatment Option for Fibrotic Diseases. J Clin Med 2023; 13:164. [PMID: 38202170 PMCID: PMC10780256 DOI: 10.3390/jcm13010164] [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: 09/29/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Fibrosis is the end result of persistent inflammatory responses induced by a variety of stimuli, including chronic infections, autoimmune reactions, and tissue injury. Fibrotic diseases affect all vital organs and are characterized by a high rate of morbidity and mortality in the developed world. Until recently, there were no approved antifibrotic therapies. In recent years, high levels of interleukin-17 (IL-17) have been associated with chronic inflammatory diseases with fibrotic complications that culminate in organ failure. In this review, we provide an update on the role of IL-17 in fibrotic diseases, with particular attention to the most recent lines of research in the therapeutic field represented by the epigenetic mechanisms that control IL-17 levels in fibrosis. A better knowledge of the IL-17 signaling pathway implications in fibrosis could design new strategies for therapeutic benefits.
Collapse
Affiliation(s)
- Margherita Sisto
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | | |
Collapse
|
46
|
Ferreira LB, Williams KA, Best G, Haydinger CD, Smith JR. Inflammatory cytokines as mediators of retinal endothelial barrier dysfunction in non-infectious uveitis. Clin Transl Immunology 2023; 12:e1479. [PMID: 38090668 PMCID: PMC10714664 DOI: 10.1002/cti2.1479] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/22/2023] [Accepted: 11/21/2023] [Indexed: 06/30/2024] Open
Abstract
Characterised by intraocular inflammation, non-infectious uveitis includes a large group of autoimmune and autoinflammatory diseases that either involve the eye alone or have both ocular and systemic manifestations. When non-infectious uveitis involves the posterior segment of the eye, specifically the retina, there is substantial risk of vision loss, often linked to breakdown of the inner blood-retinal barrier. This barrier is formed by non-fenestrated retinal vascular endothelial cells, reinforced by supporting cells that include pericytes, Müller cells and astrocytes. Across the published literature, a group of inflammatory cytokines stand out as prominent mediators of intraocular inflammation, with effects on the retinal endothelium that may contribute to breakdown of the inner blood-retinal barrier, namely tumour necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-8, IL-17 and chemokine C-C motif ligand (CCL)2. This article reviews the function of each cytokine and discusses the evidence for their involvement in retinal endothelial barrier dysfunction in non-infectious uveitis, including basic laboratory investigations, studies of ocular fluids collected from patients with non-infectious uveitis, and results of clinical treatment trials. The review also outlines gaps in knowledge in this area. Understanding the disease processes at a molecular level can suggest treatment alternatives that are directed against appropriate biological targets to protect the posterior segment of eye and preserve vision in non-infectious uveitis.
Collapse
Affiliation(s)
| | - Keryn A Williams
- Flinders University College of Medicine and Public HealthAdelaideSAAustralia
| | - Giles Best
- Flinders University College of Medicine and Public HealthAdelaideSAAustralia
| | - Cameron D Haydinger
- Flinders University College of Medicine and Public HealthAdelaideSAAustralia
| | - Justine R Smith
- Flinders University College of Medicine and Public HealthAdelaideSAAustralia
| |
Collapse
|
47
|
Hassan T, Zakaria M, Diaa A, Abdalla AELS, Ahmed ALSMS, Abdelmonem DM, Abdelaziz E. Contribution of T helper 17 cells and interleukin-17 to the pathogenesis of primary immune thrombocytopenia in Egyptian children. Eur J Pediatr 2023; 182:5673-5679. [PMID: 37823926 PMCID: PMC10746750 DOI: 10.1007/s00431-023-05242-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
Though pathogenesis of primary immune thrombocytopenia (ITP) is still rendered unclear, yet there are many research efforts that have been directed to the role of T helper 17 (Th17) and interleukin 17 (IL-17) in the pathogenesis of this disease. The Th17 cell, which produces IL-17, is a subset of T helper cells. Interleukin 17 is pro-inflammatory cytokine that is recently proved to have a crucial role in the emergence of autoimmune diseases. We aimed to investigate the role of T helper17 cells and interleukin-17 in the pathogenesis of ITP in Egyptian children. This study was carried out on 100 children with ITP and 100 apparently healthy children as a control group. Patients were subjected to full medical history taking, thorough physical examination and routine investigations according to our local standards. Percentage of Th17 cells was measured by flow cytometry in study groups. Also, serum IL-17 was measured in in study groups by ELISA. Th 17 cells were significantly higher in patients compared to controls. Moreover, 3.1-fold increased serum levels of IL-17 were observed in patients with ITP compared to controls. Newly diagnosed patients had significantly higher percentage of Th-17cells as well as higher IL-17 levels than patients with either persistent or chronic ITP. Conclusion: We concluded that Th 17 cells and IL-17 seem to play an important role in the pathogenesis of ITP in Egyptian children. What is Known -- What is New: • The pathogenesis of ITP is heterogeneous A novel subset of CD4+ T cells, distinct from Th1 and Th2, was recently identified. It is characterized by the production of IL-17 and, therefore, designated as Th17 cells. Several studies support a pivotal role for serum cytokines in the pathogenesis of ITP and provide evidence to suggest that helper Tlymphocytes polarize into Th1 and Th2 immune response. we aimed to investigate the role of T helper17 cells and interleukin-17 in the pathogenesis of ITP in Egyptian children.
Collapse
Affiliation(s)
- Tamer Hassan
- Pediatrics, Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Marwa Zakaria
- Pediatrics, Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt.
| | - Asmaa Diaa
- Pediatrics, Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | | | | | - Doaa M Abdelmonem
- Clinical Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman Abdelaziz
- Clinical Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
48
|
Zaher K, Basingab F. Interaction between Gut Microbiota and Dendritic Cells in Colorectal Cancer. Biomedicines 2023; 11:3196. [PMID: 38137417 PMCID: PMC10741039 DOI: 10.3390/biomedicines11123196] [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: 10/07/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 12/24/2023] Open
Abstract
Colorectal cancer (CRC) is a malignancy that manifests in serial stages and has been observed to have an escalating incidence in modern societies, causing a significant global health problem. The development of CRC is influenced by various exogenous factors, including lifestyle, diet, nutrition, environment, and microbiota, that can affect host cells, including immune cells. Various immune dysfunctions have been recognized in patients with CRC at different stages of this disease. The signature of microbiota in the development of CRC-inflammation related to obesity, diet, and reactive host cells, such as dendritic cells (DCs)-has been highlighted by many studies. This study focuses on DCs, the primary cellular mediators linking innate and adaptive immune responses against cancer. In addition, this review focuses on the role of microbiota in dysbiosis and how it affects DCs and, in turn, the immune response and progression of CRC by stimulating different sets of T cells. Additionally, DCs' role in protecting this delicate balance is examined. This is to determine how gene yields of commensal microbiota may be critical in restoring this balance when disrupted. The stages of the disease and major checkpoints are discussed, as well as the role of the C-type lectin receptor of immature DCs pattern recognition receptor in CRC. Finally, based on a thorough examination of worldwide clinical studies and recent advancements in cancer immunotherapy, it is recommended that innovative approaches that integrate DC vaccination strategies with checkpoint inhibitors be considered. This approach holds great promise for improving CRC management.
Collapse
Affiliation(s)
- Kawther Zaher
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Fatemah Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21585, Saudi Arabia
| |
Collapse
|
49
|
Wu Y, Luo J, Xu B. Network pharmacology and bioinformatics to identify the molecular mechanisms of Gleditsiae Spina against colorectal cancer. Curr Res Toxicol 2023; 5:100139. [PMID: 38059131 PMCID: PMC10696432 DOI: 10.1016/j.crtox.2023.100139] [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: 08/29/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023] Open
Abstract
Objective In this study, network pharmacology, bioinformatics and molecular docking were used to explore the active phytochemicals, hub genes, and potential molecular mechanisms of Gleditsiae Spina in treating of colorectal cancer.. Methods The targets of Gleditsiae Spina, and targets related to CRC were derived from databases. We identified the hub genes for Gleditsiae Spina anti-colorectal cancer following the protein-protein-interaction (PPI) network. Furthermore, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment were used to analyze the hub genes from a macro perspective. Finally, we verified the hub genes by molecular docking, GEPIA, HPA, and starBase database. Results We identified nine active phytochemicals and 36 intersection targets. The GO enrichment analysis results showed that Gleditsiae Spina may be involved in gene targets affecting multiple biological processes, including response to radiation, response to ionizing radiation, cyclin-dependent protein kinase holoenzyme complex, serine/threonine protein kinase complex, cyclin-dependent protein serine/threonine kinase regulator activity and protein kinase regulator activity. KEGG enrichment analysis results indicated that the P53 signaling pathway, IL-17 signaling pathway, Toll-like receptor signaling pathway, PI3K-Akt signaling pathway, and JAK-STAT signaling pathway were mainly related to the effect of Gleditsiae Spina on colorectal cancer. Molecular docking analysis suggested that the active phytochemicals of Gleditsiae Spina could combine well with hub genes (PTGS1, PIK3CG, CCND1, CXCL8 and ADRB2). Conclusion This study provides clues for further study of anti-CRC phytochemicals as well as their mechanisms of provides a basis for their development model.
Collapse
Affiliation(s)
- Yingzi Wu
- Guangdong Provincial Key Laboratory IRADS and Department of Life Sciences, BNU-HKBU United International College, Zhuhai 519087, China
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Jinhai Luo
- Guangdong Provincial Key Laboratory IRADS and Department of Life Sciences, BNU-HKBU United International College, Zhuhai 519087, China
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Baojun Xu
- Guangdong Provincial Key Laboratory IRADS and Department of Life Sciences, BNU-HKBU United International College, Zhuhai 519087, China
| |
Collapse
|
50
|
Garcia Moreno AS, Guicciardi ME, Wixom AQ, Jessen E, Yang J, Ilyas SI, Bianchi JK, Pinto E Vairo F, Lazaridis KN, Gores GJ. IL-17 Signaling in Primary Sclerosing Cholangitis Patient-Derived Organoids. RESEARCH SQUARE 2023:rs.3.rs-3406046. [PMID: 37886596 PMCID: PMC10602181 DOI: 10.21203/rs.3.rs-3406046/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The pathogenesis of primary sclerosing cholangitis (PSC) is unclear, although studies implicate IL-17A as an inflammatory mediator in this disease. However, a direct assessment of IL-17 signaling in PSC cholangiocytes is lacking. In this study we aimed to investigate the response of PSC extrahepatic cholangiocyte organoids (ECO) to IL-17A stimulation. Cholangiocytes obtained from PSC and non-PSC patients by endoscopic retrograde cholangiography (ERC) were cultured as ECO. The ECO were treated with vehicle or IL-17A and assessed by transcriptomics, secretome analysis, and genome sequencing (GS). Unsupervised clustering of all integrated scRNA-seq data identified 8 cholangiocyte clusters which did not differ between PSC and non-PSC ECO. However, PSC ECO cells demonstrated a robust response to IL-17 treatment, noted by an increased number of differentially expressed genes (DEG) by transcriptomics, and more abundant chemokine and cytokine expression and secretion. After rigorous filtering, GS identified candidate somatic variants shared among PSC ECO from unrelated individuals. However, no candidate rare variants in genes regulating the IL-17 pathway were identified, but rare variants regulating the MAPK signaling pathway were present in all PSC ECO. In conclusion, PSC and non-PSC patient derived ECO respond differently to IL-17 stimulation implicating this pathway in the pathogenesis of PSC.
Collapse
|