1
|
Tull S, Saviano A, Fatima A, Begum J, Mansour AA, Marigliano N, Schettino A, Blaising J, Trenkle P, Sandrin V, Maione F, Regan-Komito D, Iqbal AJ. Dichotomous effects of Galectin-9 in disease modulation in murine models of inflammatory bowel disease. Biomed Pharmacother 2025; 184:117902. [PMID: 39951917 PMCID: PMC11870847 DOI: 10.1016/j.biopha.2025.117902] [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: 09/02/2024] [Revised: 01/17/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a multifaceted disease characterised by compromised integrity of the epithelial barrier, the gut microbiome, and mucosal inflammation. While leukocyte recruitment and infiltration into intestinal tissue are well-studied and targeted in clinical practice, the role of galectins in modulating mucosal immunity remains underexplored. Galectins, a family of lectin-binding proteins, mediate critical interactions between immune cells and the intestinal epithelium. This study investigated the effect of endogenous Galectin-9 (Gal-9), as well as the combined effects with Galectin-3 (Gal-3), in modulating disease progression in murine models of colitis, using global knockout (KO) models for Gal-3, Gal-9, and Gal-3/Gal-9. Global deficiency in both galectins demonstrated improved disease parameters in Dextran sodium sulfate (DSS)-driven colitis. In contrast, in a model of adoptive T cell driven colitis, the addition of recombinant Gal-9 (rGal-9) was associated with reduced intestinal inflammation and an improvement in disease parameters. Further in vitro studies revealed no change in bone marrow-derived macrophage cytokine production in the absence of endogenous Gal-9, whereas the addition of rGal-9 to human macrophages stimulated pro-inflammatory cytokine production. Collectively, these findings demonstrate that Gal-9 plays distinct, context-dependent effects in intestinal inflammation, with both pro-inflammatory and anti-inflammatory effects. The contrasting functions of endogenous and exogenous Gal-9 underscore its complex involvement in IBD pathogenesis and highlight the need to differentiate its physiological function from therapeutic applications.
Collapse
Affiliation(s)
- Samantha Tull
- Department of Cardiovascular Sciences (CVS), College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy
| | - Areeba Fatima
- Department of Cardiovascular Sciences (CVS), College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Jenefa Begum
- Department of Cardiovascular Sciences (CVS), College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Adel Abo Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy
| | - Anna Schettino
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy
| | - Julie Blaising
- Department of Cardiovascular Sciences (CVS), College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK; ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia; Roche Pharma Research & Early Development, CMV, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Patrick Trenkle
- Roche Pharma Research & Early Development, CMV, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Virginie Sandrin
- Roche Pharma Research & Early Development, CMV, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy.
| | - Daniel Regan-Komito
- Roche Pharma Research & Early Development, CMV, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland.
| | - Asif J Iqbal
- Department of Cardiovascular Sciences (CVS), College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK; ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy.
| |
Collapse
|
2
|
Gossink EM, Coffer PJ, Cutilli A, Lindemans CA. Immunomodulation by galectin-9: Distinct role in T cell populations, current therapeutic avenues and future potential. Cell Immunol 2025; 407:104890. [PMID: 39571310 DOI: 10.1016/j.cellimm.2024.104890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 12/15/2024]
Abstract
Galectins, glycan-binding proteins, have been identified as critical regulators of the immune system. Recently, Galectin-9 (Gal-9) has emerged as biomarker that correlates with disease severity in a range of inflammatory conditions. However, Gal-9 has highly different roles in the context of immunoregulation, with the potential to either stimulate or suppress the immune response. Neutralizing antibodies targeting Gal-9 have been developed and are in early test phase investigating their therapeutic potential in cancer. Despite ongoing research, the mechanisms behind Gal-9 action remain not fully understood, and extrapolating the implications of targeting this molecule from previous studies is challenging. Here, we examine the pleiotropic function of Gal-9 focusing on conventional T lymphocytes, providing a current overview of its immunostimulatory and immunosuppressive roles. In particular, we highlight that Gal-9 differentially regulates immune responses depending on the context. Considering this complexity, further investigation of Gal-9's intricate biology is necessary to define therapeutic strategies in immune disorders and cancer treatment aimed at inducing or inhibiting Gal-9 signaling.
Collapse
Affiliation(s)
- Eva M Gossink
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, the Netherlands; Division of Pediatrics, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands; Regenerative Medicine Center, University Medical Center Utrecht, 3584CT Utrecht, the Netherlands
| | - Paul J Coffer
- Division of Pediatrics, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands; Regenerative Medicine Center, University Medical Center Utrecht, 3584CT Utrecht, the Netherlands; Center of Molecular Medicine, University Medical Center Utrecht, 3584CG Utrecht, the Netherlands
| | - Alessandro Cutilli
- Regenerative Medicine Center, University Medical Center Utrecht, 3584CT Utrecht, the Netherlands; Center of Molecular Medicine, University Medical Center Utrecht, 3584CG Utrecht, the Netherlands
| | - Caroline A Lindemans
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, the Netherlands; Division of Pediatrics, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands; Regenerative Medicine Center, University Medical Center Utrecht, 3584CT Utrecht, the Netherlands.
| |
Collapse
|
3
|
Deng J, Wu P. Integrated bioinformatics analysis and in vivo validation of potential immune-related genes linked to diabetic nephropathy. Heliyon 2024; 10:e40151. [PMID: 39583850 PMCID: PMC11582746 DOI: 10.1016/j.heliyon.2024.e40151] [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: 03/14/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024] Open
Abstract
Background Diabetic nephropathy (DN) is a common microvascular complication of diabetes mellitus and the main cause of chronic renal failure. This study explored the potential immunomodulation-related genes (IRGs) in DN using bioinformatics. Methods IRGs were identified using GeneCards, and differentially expressed genes were identified using the GSE99339, GSE96804, and GSE30122 datasets. We conducted enrichment analyses using Gene Ontology, gene set enrichment analysis (GSEA), and Kyoto Encyclopedia of Genes and Genomes to identify the associated signaling pathways. Prognostic models were constructed using Least Absolute Shrinkage and Selection Operator regression. The predictive power of IRGs was evaluated using receiver operating characteristic (ROC) curves. Furthermore, we utilized ssGSEA to determine the relative abundance of immune cell infiltration. The expression of five significant IRGs was further validated using immunohistochemistry (IHC) in individuals with DN and real-time PCR (RT-PCR) in animal experiments. Results In total, 17 immunomodulation-related differentially expressed genes were identified, which were enriched in immune-associated pathways and inflammation. GSEA unveiled substantial enrichments in metabolic irregularities and the structural composition of the extracellular matrix. ROC analysis results revealed that the diagnostic efficacy of IFNAR2 and CASP3 was comparatively high. Notably, we identified potential IRGs for DN, including CASP3, LGALS9, and SST, using IHC and RT-PCR. Conclusions CASP3, LGALS9, and SST are potential IRGs in patients with DN. Our findings may offer a theoretical basis for developing more focused and innovative immunotherapy approaches for patients with DN.
Collapse
Affiliation(s)
- Jinxiu Deng
- Department of Endocrinology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, China
| | - Peiwen Wu
- Department of Endocrinology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
| |
Collapse
|
4
|
Meng L, Chen HM, Zhang JS, Wu YR, Xu YZ. Matricellular proteins: From cardiac homeostasis to immune regulation. Biomed Pharmacother 2024; 180:117463. [PMID: 39305814 DOI: 10.1016/j.biopha.2024.117463] [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: 06/26/2024] [Revised: 08/23/2024] [Accepted: 09/19/2024] [Indexed: 11/14/2024] Open
Abstract
Tissue repair after myocardial injury is a complex process involving changes in all aspects of the myocardial tissue, including the extracellular matrix (ECM). The ECM is composed of large structural proteins such as collagen and elastin and smaller proteins with major regulatory properties called matricellular proteins. Matricellular cell proteins exert their functions and elicit cellular responses by binding to structural proteins not limited to interactions with cell surface receptors, cytokines, or proteases. At the same time, matricellular proteins act as the "bridge" of information exchange between cells and ECM, maintaining the integrity of the cardiac structure and regulating the immune environment, which is a key factor in determining cardiac homeostasis. In this review, we present an overview of the identified matricellular proteins and summarize the current knowledge regarding their roles in maintaining cardiac homeostasis and regulating the immune system.
Collapse
Affiliation(s)
- Li Meng
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, China; Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China
| | - Hui-Min Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, China; Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China
| | - Jia-Sheng Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, China; Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China
| | - Yi-Rong Wu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China.
| | - Yi-Zhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China.
| |
Collapse
|
5
|
Cao Z, Leng P, Xu H, Li X. The regulating role of galectin-9 in immune cell populations. Front Pharmacol 2024; 15:1462061. [PMID: 39539619 PMCID: PMC11557436 DOI: 10.3389/fphar.2024.1462061] [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: 07/09/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Galectin-9 (gal-9) is a protein that belongs to the galectin family. Gal-9 is expressed in cells of the innate and adaptive immune system, including lymphocytes, dendritic cells, giant salivary cells, eosinophils and T cells, etc. In different immune cells, the role of gal-9 is different. Gal-9 can induce the proliferation and activation of immune cells, and also promote the apoptosis of immune cells. This effect of gal-9 affects the occurrence and development of a variety of immune-related diseases, such as the invasion of pathogenic microorganisms, immune escape of tumor cells, and inflammatory response. Thus, understanding the biological roles of gal-9 in innate and adaptive immunity may be essential for autoimmune diseases treatment and diagnosis to improve patient quality of life. In this review, we aim to summarize current research on the regulatory roles of gal-9 in human immune system and potential inducers and inhibitors of gal-9, which may provide new strategies for immune diseases therapies.
Collapse
Affiliation(s)
- Zhanqi Cao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | | | | |
Collapse
|
6
|
Jover E, Martín-Núñez E, Garaikoetxea M, Matilla L, Blanco-Colio LM, Pérez-Sáez JM, Navarro A, Fernández-Celis A, Gainza A, Álvarez V, Sádaba R, Tamayo I, Rabinovich GA, Martín-Ventura JL, López-Andrés N. Sex-dependent expression of galectin-1, a cardioprotective β-galactoside-binding lectin, in human calcific aortic stenosis. FASEB J 2024; 38:e23447. [PMID: 38329326 DOI: 10.1096/fj.202301832rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/02/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
We aimed to analyze sex-related differences in galectin-1 (Gal-1), a β-galactoside-binding lectin, in aortic stenosis (AS) and its association with the inflammatory and fibrocalcific progression of AS. Gal-1 was determined in serum and aortic valves (AVs) from control and AS donors by western blot and immunohistochemistry. Differences were validated by ELISA and qPCR in AS samples. In vitro experiments were conducted in primary cultured valve interstitial cells (VICs). Serum Gal-1 was not different neither between control and AS nor between men and women. There was no association between circulating and valvular Gal-1 levels. The expression of Gal-1 in stenotic AVs was higher in men than women, even after adjusting for confounding factors, and was associated with inflammation, oxidative stress, extracellular matrix remodeling, fibrosis, and osteogenesis. Gal-1 (LGALS1) mRNA was enhanced within fibrocalcific areas of stenotic AVs, especially in men. Secretion of Gal-1 was up-regulated over a time course of 2, 4, and 8 days in men's calcifying VICs, only peaking at day 4 in women's VICs. In vitro, Gal-1 was associated with similar mechanisms to those in our clinical cohort. β-estradiol significantly up-regulated the activity of an LGALS1 promoter vector and the secretion of Gal-1, only in women's VICs. Supplementation with rGal-1 prevented the effects elicited by calcific challenge including the metabolic shift to glycolysis. In conclusion, Gal-1 is up-regulated in stenotic AVs and VICs from men in association with inflammation, oxidative stress, matrix remodeling, and osteogenesis. Estrogens can regulate Gal-1 expression with potential implications in post-menopause women. Exogenous rGal-1 can diminish calcific phenotypes in both women and men.
Collapse
Affiliation(s)
- Eva Jover
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Ernesto Martín-Núñez
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Mattie Garaikoetxea
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Lara Matilla
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Luis M Blanco-Colio
- IIS-Fundación Jiménez-Díaz-Autonoma University of Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan M Pérez-Sáez
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Adela Navarro
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Alicia Gainza
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Virginia Álvarez
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Rafael Sádaba
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Ibai Tamayo
- Research Methodology Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - José L Martín-Ventura
- IIS-Fundación Jiménez-Díaz-Autonoma University of Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| |
Collapse
|
7
|
Meng Y, Liu Y, Zhu Y, Qiu C, Ma A, Liu D, Zhang S, Gu L, Zhang J, Wang J. Insights into the time-course cellular effects triggered by iron oxide nanoparticles by combining proteomics with the traditional pharmacology strategy. J Mater Chem B 2024; 12:1892-1904. [PMID: 38305086 DOI: 10.1039/d3tb02476h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
In recent years, a number of initially approved magnetic iron oxide nanoparticle (IONP)-based nano-medicines have been withdrawn due to the obscure nano-bio effects. Therefore, there is an urgent need to study the cellular effects triggered by IONPs on cells. In this study, we investigate the time-course cellular effects on the response of RAW 264.7 cells caused by Si-IONPs via pharmacological and mass spectrometry-based proteomics techniques. Our results revealed that Si-IONPs were internalized by clathrin-mediated endocytosis within 1 hour, and gradually degraded in endolysosomes over time, which might influence autophagy, oxidative stress, innate immune response, and inflammatory response after 12 hours. Our research provides a necessary assessment of Si-IONPs for further clinical treatment.
Collapse
Affiliation(s)
- Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Chong Qiu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Ang Ma
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Dandan Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Shujie Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Liwei Gu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
8
|
Sun F, Cheng Y, Wanchai V, Guo W, Mery D, Xu H, Gai D, Siegel E, Bailey C, Ashby C, Al Hadidi S, Schinke C, Thanendrarajan S, Ma Y, Yi Q, Orlowski RZ, Zangari M, van Rhee F, Janz S, Bishop G, Tricot G, Shaughnessy JD, Zhan F. Bispecific BCMA/CD24 CAR-T cells control multiple myeloma growth. Nat Commun 2024; 15:615. [PMID: 38242888 PMCID: PMC10798961 DOI: 10.1038/s41467-024-44873-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
Anti-multiple myeloma B cell maturation antigen (BCMA)-specific chimeric antigen receptor (CAR) T-cell therapies represent a promising treatment strategy with high response rates in myeloma. However, durable cures following anti-BCMA CAR-T cell treatment of myeloma are rare. One potential reason is that a small subset of minimal residual myeloma cells seeds relapse. Residual myeloma cells following BCMA-CAR-T-mediated treatment show less-differentiated features and express stem-like genes, including CD24. CD24-positive myeloma cells represent a large fraction of residual myeloma cells after BCMA-CAR-T therapy. In this work, we develop CD24-CAR-T cells and test their ability to eliminate myeloma cells. We find that CD24-CAR-T cells block the CD24-Siglec-10 pathway, thereby enhancing macrophage phagocytic clearance of myeloma cells. Additionally, CD24-CAR-T cells polarize macrophages to a M1-like phenotype. A dual-targeted BCMA-CD24-CAR-T exhibits improved efficacy compared to monospecific BCMA-CAR-T-cell therapy. This work presents an immunotherapeutic approach that targets myeloma cells and promotes tumor cell clearance by macrophages.
Collapse
Affiliation(s)
- Fumou Sun
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Yan Cheng
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Visanu Wanchai
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Wancheng Guo
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - David Mery
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Hongwei Xu
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Dongzheng Gai
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Eric Siegel
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Clyde Bailey
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Cody Ashby
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Samer Al Hadidi
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Carolina Schinke
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Sharmilan Thanendrarajan
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Yupo Ma
- iCell Gene Therapeutics LLC, Research & Development Division, Stony Brook, NY, 11790, USA
| | - Qing Yi
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Maurizio Zangari
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Frits van Rhee
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Siegfried Janz
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Gail Bishop
- Department of Microbiology and Immunology, University of Iowa and VA Medical Center, Iowa City, IA, 52242, USA
| | - Guido Tricot
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - John D Shaughnessy
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Fenghuang Zhan
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
9
|
Warnakula WADLR, Udayantha HMV, Liyanage DS, Omeka WKM, Lim C, Kim G, Sirisena DMKP, Jayamali BPMV, Wan Q, Lee J. Galectin 9 restricts viral replication in teleost via autophagy-antiviral pathway and polarizes M2 macrophages for anti-inflammatory response: New insights into functional properties of fish Galectin-9 from Planiliza haematocheilus. FISH & SHELLFISH IMMUNOLOGY 2023; 143:109172. [PMID: 37858785 DOI: 10.1016/j.fsi.2023.109172] [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: 07/03/2023] [Revised: 09/27/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Galectin 9 (Gal9) is a tandem repeat type ß-galactoside-binding galectin that mediates various cellular biochemical and immunological functions. Many studies have investigated the functional properties of Gal9 in mammals; however, knowledge of fish Gal9 is limited to antibacterial studies. In this context, our aim was to clone Gal9 from Planiliza haematocheilus (PhGal9) and investigate its structural and functional characteristics. We discovered the PhGal9 open reading frame, which was 969 base pairs long and encoded a 322 amino acid protein. PhGal9 had a projected molecular weight of 35.385 kDa but no signal peptide sequence. PhGal9 mRNA was ubiquitously produced in all investigated tissues but was predominant in the intestine, spleen, and brain. Its mRNA expression was increased in response to stimulation by Poly(I:C), LPS, and L. garvieae. The rPhGal9 exhibited a dose-dependent agglutination potential toward gram-positive and gram-negative bacteria at a minimum concentration of 50 μg/mL. Overexpression of PhGal9 promoted M2-like phenotype changes in mouse macrophages, and RT-qPCR analysis of M1 and M2 marker genes confirmed M2 polarization with upregulation of M2 marker genes. In the antiviral assay, the expression levels of Viral Hemorrhagic Septicemia Virus (VHSV) glycoproteins, phosphoproteins, nucleoproteins, non-virion proteins, matrix proteins, and RNA polymerase were significantly reduced in PhGal9-overexpressed cells. Furthermore, the mRNA expression of autophagic genes (sqstm1, tax1bp1b, rnf13, lc3, and atg5) and antiviral genes (viperin) were upregulated in PhGal9 overexpressed cells. For the first time in teleosts, our study demonstrated that PhGal9 promotes M2 macrophage polarization by upregulating M2-associated genes (egr2 and cmyc) and suppressing M1-associated genes (iNOS and IL-6). Furthermore, our results show that exogenous and endogenous PhGal9 prevented VHSV attachment and replication by neutralizing virion and autophagy, respectively. Gal9 may be a potent modulator of the antimicrobial immune response in teleost fish.
Collapse
Affiliation(s)
- W A D L R Warnakula
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - H M V Udayantha
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju, 63333, Republic of Korea
| | - D S Liyanage
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju, 63333, Republic of Korea
| | - W K M Omeka
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju, 63333, Republic of Korea
| | - Chaehyeon Lim
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Gaeun Kim
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - D M K P Sirisena
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - B P M Vileka Jayamali
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Qiang Wan
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju, 63333, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju, 63333, Republic of Korea.
| |
Collapse
|
10
|
Biagioli M, Marchianò S, Di Giorgio C, Bordoni M, Urbani G, Bellini R, Massa C, Sami Ullah Khan R, Roselli R, Chiara Monti M, Morretta E, Giordano A, Vellecco V, Bucci M, Jilani Iqbal A, Saviano A, Ab Mansour A, Ricci P, Distrutti E, Zampella A, Cieri E, Cirino G, Fiorucci S. Activation of GPBAR1 attenuates vascular inflammation and atherosclerosis in a mouse model of NAFLD-related cardiovascular disease. Biochem Pharmacol 2023; 218:115900. [PMID: 37926268 DOI: 10.1016/j.bcp.2023.115900] [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: 08/29/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
While patients with nonalcoholic fatty liver disease (NAFLD) are at increased risk to develop clinically meaningful cardiovascular diseases (CVD), there are no approved drug designed to target the liver and CVD component of NAFLD. GPBAR1, also known as TGR5, is a G protein coupled receptor for secondary bile acids. In this study we have investigated the effect of GPBAR1 activation by BAR501, a selective GPBAR1 agonist, in Apolipoprotein E deficient (ApoE-/-) mice fed a high fat diet and fructose (Western diet), a validated model of NAFLD-associated atherosclerosis. Using aortic samples from patients who underwent surgery for abdominal aneurism, and ex vivo experiments with endothelial cells and human macrophages, we were able to co-localize the expression of GPBAR1 in CD14+ and PECAM1+ cells. Similar findings were observed in the aortic plaques from ApoE-/- mice. Treating ApoE-/- mice with BAR501, 30 mg/kg for 14 weeks, attenuated the body weight gain while ameliorated the insulin sensitivity by increasing the plasma concentrations of GLP-1 and FGF15. Activation of GPBAR1 reduced the aorta thickness and severity of atherosclerotic lesions and decreased the amount of plaques macrophages. Treating ApoE-/- mice reshaped the aortic transcriptome promoting the expression of anti-inflammatory genes, including IL-10, as also confirmed by tSNE analysis of spleen-derived macrophages. Feeding ApoE-/- mice with BAR501 redirected the bile acid synthesis and the composition of the intestinal microbiota. In conclusion, GPBAR1 agonism attenuates systemic inflammation and improve metabolic profile in a genetic/dietetic model of atherosclerosis. BAR501 might be of utility in the treatment for NAFLD-related CVD.
Collapse
Affiliation(s)
- Michele Biagioli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Silvia Marchianò
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Martina Bordoni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Ginevra Urbani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rachele Bellini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Carmen Massa
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Rosalinda Roselli
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Elva Morretta
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Antonino Giordano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | | | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Anella Saviano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Adel Ab Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Patrizia Ricci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Angela Zampella
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Enrico Cieri
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giuseppe Cirino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Stefano Fiorucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
11
|
Saviano A, Manosour AA, Raucci F, Merlino F, Marigliano N, Schettino A, Wahid M, Begum J, Filer A, Manning JE, Casillo GM, Piccolo M, Ferraro MG, Marzano S, Russomanno P, Bellavita R, Irace C, Amato J, Alfaifi M, Rimmer P, Iqbal T, Pieretti S, Vellecco V, Caso F, Costa L, Giacomelli R, Scarpa R, Cirino G, Bucci M, McGettrick HM, Grieco P, Iqbal AJ, Maione F. New biologic (Ab-IPL-IL-17) for IL-17-mediated diseases: identification of the bioactive sequence (nIL-17) for IL-17A/F function. Ann Rheum Dis 2023; 82:1415-1428. [PMID: 37580108 PMCID: PMC10579190 DOI: 10.1136/ard-2023-224479] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/07/2023] [Indexed: 08/16/2023]
Abstract
OBJECTIVES Interleukin (IL) 17s cytokines are key drivers of inflammation that are functionally dysregulated in several human immune-mediated inflammatory diseases (IMIDs), such as rheumatoid arthritis (RA), psoriasis and inflammatory bowel disease (IBD). Targeting these cytokines has some therapeutic benefits, but issues associated with low therapeutic efficacy and immunogenicity for subgroups of patients or IMIDs reduce their clinical use. Therefore, there is an urgent need to improve the coverage and efficacy of antibodies targeting IL-17A and/or IL-17F and IL-17A/F heterodimer. METHODS AND RESULTS Here, we initially identified a bioactive 20 amino acid IL-17A/F-derived peptide (nIL-17) that mimics the pro-inflammatory actions of the full-length proteins. Subsequently, we generated a novel anti-IL-17 neutralising monoclonal antibody (Ab-IPL-IL-17) capable of effectively reversing the pro-inflammatory, pro-migratory actions of both nIL-17 and IL-17A/F. Importantly, we demonstrated that Ab-IPL-IL-17 has less off-target effects than the current gold-standard biologic, secukinumab. Finally, we compared the therapeutic efficacy of Ab-IPL-IL-17 with reference anti-IL-17 antibodies in preclinical murine models and samples from patients with RA and IBD. We found that Ab-IPL-IL-17 could effectively reduce clinical signs of arthritis and neutralise elevated IL-17 levels in IBD patient serum. CONCLUSIONS Collectively, our preclinical and in vitro clinical evidence indicates high efficacy and therapeutic potency of Ab-IPL-IL-17, supporting the rationale for large-scale clinical evaluation of Ab-IPL-IL-17 in patients with IMIDs.
Collapse
Affiliation(s)
- Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Adel Abo Manosour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Francesco Merlino
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Anna Schettino
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Mussarat Wahid
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jenefa Begum
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrew Filer
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Julia E Manning
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | - Marialuisa Piccolo
- BioChemLab, Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Maria Grazia Ferraro
- BioChemLab, Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Simona Marzano
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | | | - Rosa Bellavita
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Carlo Irace
- BioChemLab, Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Mohammed Alfaifi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Peter Rimmer
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Gastroenterology, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Tariq Iqbal
- Department of Gastroenterology, Queen Elizabeth Hospital Birmingham, Birmingham, UK
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Stefano Pieretti
- Department of Drug Research and Evaluation, Istituto Superiore di Sanità, Roma, Italy
| | | | - Francesco Caso
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Luisa Costa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Roberto Giacomelli
- Fondazione Policlinico Universitario, and Research Unit of Immuno-Rheumatology, Department of Medicine and Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Roma, Italy, and Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy, Roma, Italy
| | - Raffaele Scarpa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Giuseppe Cirino
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | | | - Helen M McGettrick
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Paolo Grieco
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Asif Jilani Iqbal
- Department of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| |
Collapse
|
12
|
Lv Y, Ma X, Ma Y, Du Y, Feng J. A new emerging target in cancer immunotherapy: Galectin-9 (LGALS9). Genes Dis 2023; 10:2366-2382. [PMID: 37554219 PMCID: PMC10404877 DOI: 10.1016/j.gendis.2022.05.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/09/2022] [Accepted: 05/14/2022] [Indexed: 11/20/2022] Open
Abstract
Over the past few decades, advances in immunological knowledge have led to the identification of novel immune checkpoints, reinvigorating cancer immunotherapy. Immunotherapy, represented by immune checkpoint inhibitors, has become the leader in the precision treatment of cancer, bringing a new dawn to the treatment of most cancer patients. Galectin-9 (LGALS9), a member of the galectin family, is a widely expressed protein involved in immune regulation and tumor pathogenesis, and affects the prognosis of various types of cancer. Galectin-9 regulates immune homeostasis and tumor cell survival through its interaction with its receptor Tim-3. In the review, based on a brief description of the signaling mechanisms and immunomodulatory activities of galectin-9 and Tim-3, we summarize the targeted expression patterns of galectin-9 in a variety of malignancies and the promising mechanisms of anti-galectin-9 therapy in stimulating anti-tumor immune responses.
Collapse
Affiliation(s)
- Yan Lv
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Xiao Ma
- Department of General Surgery, The Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu 210009, China
| | - Yuxin Ma
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Yuxin Du
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Jifeng Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| |
Collapse
|
13
|
Xiong H, Xue G, Zhang Y, Wu S, Zhao Q, Zhao R, Zhou N, Xie Y. Effect of exogenous galectin-9, a natural TIM-3 ligand, on the severity of TNBS- and DSS-induced colitis in mice. Int Immunopharmacol 2023; 115:109645. [PMID: 36610329 DOI: 10.1016/j.intimp.2022.109645] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023]
Abstract
Inflammatory bowel disease (IBD) have a complex pathogenesis that is yet to be completely understood. However, a strong correlation between Toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signaling and IBD has been observed. T-cell immunoglobulin and mucin domain-containing-3 (Tim-3) has been reported to regulate TLR4/NF-κB by interacting with Galectin-9 (Gal-9), and recombinant Gal-9 can activate Tim-3; however, its potential properties in IBD and the underlying mechanism remain unclear. This study aimed to determine how Gal-9 affects experimental colitis in mice. Dextran sodium sulfate (DSS) and 2,4,6-trinitrobenzene sulfonic acid (TNBS) were used to establish colitis in mice, and the severity of the illness was assessed based on body weight, colon length, and histology. Therefore, we explored the effects of Gal-9 treatment on colitis. Furthermore, we analyzed the effect of Gal-9 on the expression of Tim-3 and TLR4/NF-κB pathway in colonic tissues and the serum levels of interferon-gamma (IFN-γ), interleukin (IL)-1β, and IL-6. Tim-3 expression in the colon was notably decreased in mice with TNBS-induced colitis, whereas TLR4/NF-kB expression was significantly increased. Intraperitoneal injection of Gal-9 dramatically decreased the disease activity index and attenuated the level of intestinal mucosal inflammation in TNBS-induced colitis mice (p < 0.05). Intraperitoneal administration of Gal-9 significantly increased Tim-3 expression in the colon and decreased the serum concentrations of IFN-γ, IL-1β, and IL-6. Additionally, Gal-9 treatment significantly downregulated the expression of TLR4 signaling pathway-related proteins. In contrast, Gal-9 did not reduce the severity of DSS-induced colitis. In summary, exogenous Gal-9 increased Tim-3 expression, inhibited the TLR4/NF-κB pathway, and alleviated TNBS-induced colitis in mice but not DSS-induced colitis in mice, revealing its potential therapeutic ramifications for IBD.
Collapse
Affiliation(s)
- Huifang Xiong
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province 330006, China; Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi 330006, China; JiangXi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China
| | - Guohui Xue
- Department of Clinical Laboratory, Jiujiang NO.1 People's Hospital, Jiujiang, Jiangxi 332000, China
| | - Yuting Zhang
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province 330006, China; Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi 330006, China; JiangXi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China
| | - Shuang Wu
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province 330006, China; Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi 330006, China; JiangXi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China
| | - Qiaoyun Zhao
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province 330006, China; Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi 330006, China; JiangXi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China
| | - Rulin Zhao
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province 330006, China; Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi 330006, China; JiangXi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China
| | - Nanjin Zhou
- Jiangxi Provincial Academy of Medical Science, Nanchang, Jiangxi 330006, China
| | - Yong Xie
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province 330006, China; Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi 330006, China; JiangXi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
14
|
Tsai MT, Yang RB, Ou SM, Tseng WC, Lee KH, Yang CY, Chang FP, Tarng DC. Plasma Galectin-9 Is a Useful Biomarker for Predicting Renal Function in Patients Undergoing Native Kidney Biopsy. Arch Pathol Lab Med 2023; 147:167-176. [PMID: 35687787 DOI: 10.5858/arpa.2021-0466-oa] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 02/05/2023]
Abstract
CONTEXT.— Galectin-9 reduces tissue damage in certain immune-mediated glomerular diseases. However, its role in structural and functional renal changes in patients with varying types of chronic kidney disease (CKD) is less clear. OBJECTIVE.— To investigate the association between plasma galectin-9 levels, proteinuria, tubulointerstitial lesions, and renal function in different CKD stages. DESIGN.— We measured plasma galectin-9 levels in 243 patients undergoing renal biopsy for determining the CKD etiology. mRNA and protein expression levels of intrarenal galectin-9 were assessed by quantitative real-time polymerase chain reaction and immunostaining. Relationships between plasma galectin-9, clinical characteristics, and tubulointerstitial damage were analyzed with logistic regression. We investigated galectin-9 expression patterns in vitro in murine J774 macrophages treated with differing stimuli. RESULTS.— To analyze the relationship between galectin-9 and clinical features, we divided the patients into 2 groups according to median plasma galectin-9 levels. The high galectin-9 group tended to be older and to have decreased renal function, higher proteinuria, and greater interstitial fibrosis. After multivariable adjustment, elevated plasma galectin-9 levels were independently associated with stage 3b or higher CKD. An analysis of gene expression in the tubulointerstitial compartment in the biopsy samples showed a significant positive correlation between intrarenal galectin-9 mRNA expression and plasma galectin-9 levels. Immunohistochemistry confirmed increased galectin-9 expression in the renal interstitium of patients with advanced CKD, and most galectin-9-positive cells were macrophages, as determined by double-immunofluorescence staining. In vitro experiments showed that galectin-9 expression in macrophages was significantly increased after interferon-γ stimulation. CONCLUSIONS.— Our findings suggest that plasma galectin-9 is a good biomarker for diagnosing advanced CKD.
Collapse
Affiliation(s)
- Ming-Tsun Tsai
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan.,Tsai and R-B Yang contributed equally to this manuscript
| | - Ruey-Bing Yang
- From the Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (R-B Yang).,Tsai and R-B Yang contributed equally to this manuscript
| | - Shuo-Ming Ou
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Cheng Tseng
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kuo-Hua Lee
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Yu Yang
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fu-Pang Chang
- From the Department of Pathology and Laboratory Medicine (Chang), Taipei Veterans General Hospital, Taipei, Taiwan
| | - Der-Cherng Tarng
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan.,From the Department and Institute of Physiology (Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
15
|
Progress of LPS-induced apical lesion in rat immature mandibular molars. PEDIATRIC DENTAL JOURNAL 2023. [DOI: 10.1016/j.pdj.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
16
|
Comparison of the Single-Cell Immune Landscape of Testudines from Different Habitats. Cells 2022; 11:cells11244023. [PMID: 36552787 PMCID: PMC9816942 DOI: 10.3390/cells11244023] [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: 10/08/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Testudines, also known as living fossils, have evolved diversely and comprise many species that occupy a variety of ecological niches. However, the immune adaptation of testudines to the different ecological niches remains poorly understood. This study compared the composition, function, and differentiation trajectories of peripheral immune cells in testudines (Chelonia mydas, Trachemys scripta elegans, Chelonoidis carbonaria, and Pelodiscus sinensis) from different habitats using the single-cell RNA sequencing (scRNA-seq) technique. The results showed that T. scripta elegans, which inhabits freshwater and brackish environments, had the most complex composition of peripheral immune cells, with 11 distinct immune cell subsets identified in total. The sea turtle C. mydas, had the simplest composition of peripheral immune cells, with only 5 distinct immune cell clusters. Surprisingly, neither basophils were found in C. mydas nor T cells in C. carbonaria. Basophil subsets in peripheral blood were identified for the first time; two basophil subtypes (GATA2-high-basophils and GATA2-low-basophils) were observed in the peripheral blood of T. scripta elegans. In addition, ACKR4 cells, CD4 T cells, CD7 T cells, serotriflin cells, and ficolin cells were specifically identified in the peripheral blood of T. scripta elegans. Furthermore, LY6G6C cells, SPC24 cells, and NKT cells were specifically observed in C. carbonaria. Moreover, there were differences in the functional status and developmental trajectory of peripheral immune cells among the testudine species. The identification of specific features of peripheral immune cells in testudines from different habitats may enable elucidation of the adaptation mechanism of testudines to various ecological niches.
Collapse
|
17
|
Krautter F, Hussain MT, Zhi Z, Lezama DR, Manning JE, Brown E, Marigliano N, Raucci F, Recio C, Chimen M, Maione F, Tiwari A, McGettrick HM, Cooper D, Fisher EA, Iqbal AJ. Galectin-9: A novel promoter of atherosclerosis progression. Atherosclerosis 2022; 363:57-68. [PMID: 36459823 DOI: 10.1016/j.atherosclerosis.2022.11.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis is widely accepted to be an inflammatory disease driven by lipid accumulation and leukocyte recruitment. More recently, galectins, a family of β-galactoside binding proteins, have been shown to play a role in leukocyte recruitment among other immunomodulatory functions. Galectin (Gal) -9, a tandem repeat type galectin expressed by the endothelium in inflammatory environments, has been proposed to promote leukocyte recruitment. However, the role of Gal-9 in the context of monocyte recruitment remains elusive. METHODS AND RESULTS Here, we characterise the immunomodulatory role of Gal-9 in context of atherosclerosis. We show that ApoE-/-Gal-9-/- mice have a significantly reduced aortic plaque burden compared to their ApoE-/- littermate controls after 12 weeks of high fat diet. RNA sequencing data from two independent studies reveal Lgals9 expression in leukocyte clusters isolated from murine atherosclerotic plaques. Additionally, soluble Gal-9 protein induces monocyte activation and a pro-inflammatory phenotype in macrophages. Furthermore, we show that immobilised recombinant Gal-9 acts as capture and adhesion molecule for CD14+ monocytes in a β2-integrin and glycan dependent manner, while adhesion of monocytes to stimulated endothelium is reduced when Gal-9 is knocked down. Gal-9 also facilitates enhanced recruitment of leukocytes from peripheral arterial disease (PAD) patients compared to healthy young and aged controls. We further characterise the endothelium as source of circulating Gal-9, which is increased in plasma of PAD patients compared to healthy controls. CONCLUSIONS These results highlight a pathological role for Gal-9 as promoter of monocyte recruitment and atherosclerotic plaque progression, making it a novel target in the prevention of plaque formation and progression.
Collapse
Affiliation(s)
- Franziska Krautter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Mohammed T Hussain
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom; The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Zhaogong Zhi
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Danielle R Lezama
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Julia E Manning
- Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Emily Brown
- Department of Medicine, Division of Cardiology, And the Cardiovascular Research Center, NYU School of Medicine, New York, United States
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carlota Recio
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Farmacología Molecular y Translacional - BIOPharm, Las Palmas de G.C, Spain
| | - Myriam Chimen
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Alok Tiwari
- Department of Vascular Surgery, University Hospitals Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Helen M McGettrick
- Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dianne Cooper
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, And the Cardiovascular Research Center, NYU School of Medicine, New York, United States
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom; ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
18
|
Mansour AA, Raucci F, Sevim M, Saviano A, Begum J, Zhi Z, Pezhman L, Tull S, Maione F, Iqbal AJ. Galectin-9 supports primary T cell transendothelial migration in a glycan and integrin dependent manner. Biomed Pharmacother 2022; 151:113171. [PMID: 35643073 DOI: 10.1016/j.biopha.2022.113171] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 05/22/2022] [Indexed: 11/26/2022] Open
Abstract
Adaptive immunity relies on the efficient recruitment of T cells from the blood into peripheral tissues. However, the current understanding of factor(s) coordinating these events is incomplete. Previous studies on galectin-9 (Gal-9), have proposed a functionally significant role for this lectin in mediating leukocyte adhesion and transmigration. However, very little is known about its function in T cell migration. Here, we have investigated the role of the Gal-9 on the migration behaviour of both human primary CD4+ and CD8+ T cells. Our data indicate that Gal-9 supports both CD4+ and CD8+ T cell adhesion and transmigration in a glycan dependent manner, inducing L-selectin shedding and upregulation of LFA-1 and CXCR4 expression. Additionally, when immobilized, Gal-9 promoted capture and firm adhesion of T cells under flow, in a glycan and integrin-dependent manner. Using an in vivo model, dorsal air pouch, we found that Gal-9 deficient mice display impaired leukocyte trafficking, with a reduction in pro-inflammatory cytokines/chemokines generated locally. Furthermore, we also demonstrate that Gal-9 inhibits the chemotactic function of CXCL12 through direct binding. In conclusion, our study characterises, for the first time, the capture, adhesion, and migration behaviour of CD4+ and CD8+ T cells to immobilised /endothelial presented Gal-9, under static and physiological flow conditions. We also demonstrate the differential binding characteristics of Gal-9 to T cell subtypes, which could be of potential therapeutic significance, particularly in the treatment of inflammatory-based diseases, given Gal-9 ability to promote apoptosis in pathogenic T cell subsets.
Collapse
Affiliation(s)
- Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Mustafa Sevim
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom; Physiology Department, School of Medicine, Marmara University, İstanbul, Turkey
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Jenefa Begum
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Zhaogong Zhi
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Laleh Pezhman
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Samantha Tull
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom; ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
19
|
Galectin-9 Triggers Neutrophil-Mediated Anticancer Immunity. Biomedicines 2021; 10:biomedicines10010066. [PMID: 35052746 PMCID: PMC8772786 DOI: 10.3390/biomedicines10010066] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022] Open
Abstract
In earlier studies, galectin-9 (Gal-9) was identified as a multifaceted player in both adaptive and innate immunity. Further, Gal-9 had direct cytotoxic and tumor-selective activity towards cancer cell lines of various origins. In the current study, we identified that treatment with Gal-9 triggered pronounced membrane alterations in cancer cells. Specifically, phosphatidyl serine (PS) was rapidly externalized, and the anti-phagocytic regulator, CD47, was downregulated within minutes. In line with this, treatment of mixed neutrophil/tumor cell cultures with Gal-9 triggered trogocytosis and augmented antibody-dependent cellular phagocytosis of cancer cells. Interestingly, this pro-trogocytic effect was also due to the Gal-9-mediated activation of neutrophils with upregulation of adhesion markers and mobilization of gelatinase, secretory, and specific granules. These activation events were accompanied by a decrease in cancer cell adhesion in mixed cultures of leukocytes and cancer cells. Further, prominent cytotoxicity was detected when leukocytes were mixed with pre-adhered cancer cells, which was abrogated when neutrophils were depleted. Taken together, Gal-9 treatment potently activated neutrophil-mediated anticancer immunity, resulting in the elimination of epithelial cancer cells.
Collapse
|
20
|
Lightfoot A, McGettrick HM, Iqbal AJ. Vascular Endothelial Galectins in Leukocyte Trafficking. Front Immunol 2021; 12:687711. [PMID: 34140956 PMCID: PMC8204101 DOI: 10.3389/fimmu.2021.687711] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/12/2021] [Indexed: 12/16/2022] Open
Abstract
Leukocyte recruitment to the site of injury is a crucial event in the regulation of an inflammatory response. Tight regulation of interactions between the endothelium and circulating leukocytes is necessary to ensure a protective response to injury does not result in inflammatory disease. Rising interest in the broad immunoregulatory roles displayed by members of the glycan-binding galectin family suggests that these proteins could be an attractive target for therapeutic intervention, since their expression is significantly altered in disease. The focus of this review is to summarize current knowledge on the role of galectins in leukocyte trafficking during inflammation and the clinical approaches being taken to target these interactions for treatment of inflammatory disease.
Collapse
Affiliation(s)
- Abbey Lightfoot
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Helen M McGettrick
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
21
|
Liang L, Zhang YM, Shen YW, Song AP, Li WL, Ye LF, Lu X, Wang GC, Peng QL. Aberrantly Expressed Galectin-9 Is Involved in the Immunopathogenesis of Anti-MDA5-Positive Dermatomyositis-Associated Interstitial Lung Disease. Front Cell Dev Biol 2021; 9:628128. [PMID: 33842457 PMCID: PMC8027128 DOI: 10.3389/fcell.2021.628128] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/04/2021] [Indexed: 01/23/2023] Open
Abstract
Background Dermatomyositis (DM) associated rapidly progressive interstitial lung disease (RP-ILD) has high mortality rate and poor prognosis. Galectin-9 (Gal-9) plays multiple functions in immune regulation. We investigated Gal-9 expression in DM patients and its association with DM-ILD. Methods A total of 154 idiopathic inflammatory myopathy patients and 30 healthy controls were enrolled in the study. Cross-sectional and longitudinal studies were used to analyze the association between serum Gal-9 levels and clinical features. Enzyme-linked immunosorbent assay and qRT-PCR were used to examine Gal-9 expression in the sera and isolated peripheral blood mononuclear cells (PBMCs) from DM patients. Immunohistochemistry was performed to analyze the expression of Gal-9 and its ligand (T-cell immunoglobulin mucin (Tim)-3 and CD44) in lung tissues from anti-melanoma differentiation-associated gene 5 (MDA5)-positive patients. The effect of Gal-9 on human lung fibroblasts (MRC-5) was investigated in vitro. Results Serum Gal-9 levels were significantly higher in DM patients than in immune-mediated necrotizing myopathy patients and healthy controls (all p < 0.001). Higher serum Gal-9 levels were observed in anti-MDA5-positive DM patients than in anti-MDA5-negative DM patients [33.8 (21.9–44.7) vs. 16.2 (10.0–26.9) ng/mL, p < 0.001]. Among the anti-MDA5-positive DM patients, serum Gal-9 levels were associated with RP-ILD severity. Serum Gal-9 levels were significantly correlated with disease activity in anti-MDA5-positive DM patients in both cross-sectional and longitudinal studies. PBMCs isolated from anti-MDA5-positive DM patients (3.7 ± 2.3 ng/mL) produced higher levels of Gal-9 than those from immune-mediated necrotizing myopathy patients (1.1 ± 0.3 ng/mL, p = 0.022) and healthy controls (1.4 ± 1.2 ng/mL, p = 0.045). The mRNA levels of Gal-9 were positively correlated with the levels of type-I interferon-inducible genes MX1 (r = 0.659, p = 0.020) and IFIH1 (r = 0.787, p = 0.002) in PBMCs from anti-MDA5-positive DM patients. Immunohistochemistry revealed increased Gal-9 and Tim-3 expression in the lung tissues of patients with DM and RP-ILD. In vitro stimulation with Gal-9 protein increased CCL2 mRNA expression in MRC-5 fibroblasts. Conclusions Among anti-MDA5-positive DM patients, Gal-9 could be a promising biomarker for monitoring disease activity, particularly for RP-ILD severity. Aberrant expression of the Gal-9/Tim-3 axis may be involved in the immunopathogenesis of DM-ILD.
Collapse
Affiliation(s)
- Lin Liang
- Department of Rheumatology, Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Ya-Mei Zhang
- Department of Rheumatology, Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Ya-Wen Shen
- Department of Rheumatology, Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Ai-Ping Song
- Department of Pathology, China-Japan Friendship Hospital, Beijing, China
| | - Wen-Li Li
- Department of Rheumatology, Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Li-Fang Ye
- Department of Rheumatology, Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Xin Lu
- Department of Rheumatology, Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Guo-Chun Wang
- Department of Rheumatology, Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Qing-Lin Peng
- Department of Rheumatology, Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
22
|
Cutine AM, Bach CA, Veigas F, Merlo JP, Laporte L, Manselle Cocco MN, Massaro M, Sarbia N, Perrotta RM, Mahmoud YD, Rabinovich GA. Tissue-specific control of galectin-1-driven circuits during inflammatory responses. Glycobiology 2021; 31:891-907. [PMID: 33498084 DOI: 10.1093/glycob/cwab007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/12/2020] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
The relevance of glycan-binding protein in immune tolerance and inflammation has been well established, mainly by studies of C-type lectins, siglecs and galectins both in experimental models and patient samples. Galectins, a family of evolutionarily conserved lectins, are characterized by sequence homology in the carbohydrate-recognition domain (CRD), atypical secretion via an ER-Golgi-independent pathway and the ability to recognize β-galactoside-containing saccharides. Galectin-1 (Gal-1), a prototype member of this family displays mainly anti-inflammatory and immunosuppressive activities, although, similar to many cytokines and growth factors, it may also trigger paradoxical pro-inflammatory effects under certain circumstances. These dual effects could be associated to tissue-, time- or context-dependent regulation of galectin expression and function, including particular pathophysiologic settings and/or environmental conditions influencing the structure of this lectin, as well as the availability of glycosylated ligands in immune cells during the course of inflammatory responses. Here, we discuss the tissue-specific role of Gal-1 as a master regulator of inflammatory responses across different pathophysiologic settings, highlighting its potential role as a therapeutic target. Further studies designed at analyzing the intrinsic and extrinsic pathways that control Gal-1 expression and function in different tissue microenvironments may contribute to design tailored therapeutic strategies aimed at positively or negatively modulate this glycan-binding protein in pathologic inflammatory conditions.
Collapse
Affiliation(s)
- Anabela M Cutine
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Camila A Bach
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Florencia Veigas
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Joaquín P Merlo
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Lorena Laporte
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Montana N Manselle Cocco
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Mora Massaro
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Nicolas Sarbia
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Ramiro M Perrotta
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Yamil D Mahmoud
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Instituto de Biología y Medicina Experimental (IBYME), Laboratorios de Inmunopatología, Glicómica Funcional e Inmuno-Oncología Translacional, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428 Buenos Aires, Argentina
| |
Collapse
|