1
|
Rosati D, Valentine M, Bruno M, Pradhan A, Dietschmann A, Jaeger M, Leaves I, van de Veerdonk FL, Joosten LA, Roy S, Stappers MHT, Gow NA, Hube B, Brown AJ, Gresnigt MS, Netea MG. Lactic acid in the vaginal milieu modulates the Candida-host interaction. Virulence 2025; 16:2451165. [PMID: 39843417 PMCID: PMC11760238 DOI: 10.1080/21505594.2025.2451165] [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: 08/19/2024] [Revised: 11/07/2024] [Accepted: 12/28/2024] [Indexed: 01/24/2025] Open
Abstract
Vulvovaginal candidiasis (VVC) is one of the most common infections caused by Candida albicans. VVC is characterized by an inadequate hyperinflammatory response and clinical symptoms associated with Candida colonization of the vaginal mucosa. Compared to other host niches in which C. albicans can cause infection, the vaginal environment is extremely rich in lactic acid that is produced by the vaginal microbiota. We examined how lactic acid abundance in the vaginal niche impacts the interaction between C. albicans and the human immune system using an in vitro culture in vaginal simulative medium (VSM). The presence of lactic acid in VSM (VSM+LA) increased C. albicans proliferation, hyphal length, and its ability to cause damage during subsequent infection of vaginal epithelial cells. The cell wall of C. albicans cells grown in VSM+LA displayed a robust mannan fibrillar structure, β-glucan exposure, and low chitin content. These cell wall changes were associated with altered immune responses and an increased ability of the fungus to induce trained immunity. Neutrophils were compromised in clearing C. albicans grown in VSM+LA conditions, despite mounting stronger oxidative responses. Collectively, we found that fungal adaptation to lactic acid in a vaginal simulative context increases its immunogenicity favouring a pro-inflammatory state. This potentially contributes to the immune response dysregulation and neutrophil recruitment observed during recurrent VVC.
Collapse
Affiliation(s)
- Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
| | - Marisa Valentine
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany
| | - Mariolina Bruno
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
| | - Arnab Pradhan
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany
| | - Martin Jaeger
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
| | - Ian Leaves
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sumita Roy
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Mark H. T. Stappers
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Neil A.R. Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany
| | - Alistair J.P. Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Mark S. Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
2
|
Zhou CK, Liu ZZ, Peng ZR, Luo XY, Zhang XM, Zhang JG, Zhang L, Chen W, Yang YJ. M28 family peptidase derived from Peribacillus frigoritolerans initiates trained immunity to prevent MRSA via the complosome-phosphatidylcholine axis. Gut Microbes 2025; 17:2484386. [PMID: 40159598 PMCID: PMC11959922 DOI: 10.1080/19490976.2025.2484386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a major global health threat due to its resistance to conventional antibiotics. The commensal microbiota maintains a symbiotic relationship with the host, playing essential roles in metabolism, energy regulation, immune modulation, and pathogen control. Mammals harbor a wide range of commensal bacteria capable of producing unique metabolites with potential therapeutic properties. This study demonstrated that M28 family peptidase (M28), derived from commensal bacteria Peribacillus frigoritolerans (P. f), provided protective effects against MRSA-induced pneumonia. M28 enhanced the phagocytosis and bactericidal activity of macrophages by inducing trained immunity. RNA sequencing and metabolomic analyses identified the CFB-C3a-C3aR-HIF-1α axis-mediated phosphatidylcholine accumulation as the key mechanism for M28-induced trained immunity. Phosphatidylcholine, like M28, also induced trained immunity. To enhance M28-mediated therapeutic potential, it was encapsulated in liposomes (M28-LNPs), which exhibited superior immune-stimulating properties compared to M28 alone. In vivo experiments revealed that M28-LNPs significantly reduced bacterial loads and lung damage following MRSA infection, which also provided enhanced protection against Klebsiella pneumoniae and Candida albicans. We first confirmed a link between complement activation and trained immunity, offering valuable insights into the treatment and prevention of complement-related autoimmune diseases.
Collapse
Affiliation(s)
- Cheng-Kai Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Zhen-Zhen Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Zi-Ran Peng
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Xue-Yue Luo
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Xiao-Mei Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Jian-Gang Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Liang Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Wei Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Yong-Jun Yang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| |
Collapse
|
3
|
Mishra A, Rawat V, Zhang K, Jagannath C. The pathway of autophagy in the epigenetic landscape of Mycobacterium-host interactions. Autophagy 2025. [PMID: 40413755 DOI: 10.1080/15548627.2025.2511074] [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: 12/04/2024] [Revised: 05/18/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025] Open
Abstract
Macroautophagy (autophagy) is an evolutionarily conserved process that degrades excess cytoplasmic components, such as protein aggregates and damaged organelles, by encapsulating them within double-membrane autophagosomes. These autophagosomes undergo distinct stages - initiation, phagophore nucleation, expansion, and closure - before fusing with lysosomes (or occasionally endosomes) for degradation and recycling. This process is regulated by ATG (autophagy related) proteins, which govern autophagosome formation and lysosomal fusion. Epigenetic modifications and transcription factors can regulate ATG gene expression in the nucleus. Autophagy also plays a key role in eliminating intracellular Mycobacterium tuberculosis (Mtb) through the lytic and antimicrobial activities of autolysosomes, which are more potent antimicrobial compartments than conventional phagosomes. Emerging evidence suggests that Mtb can modify the host epigenome and transcriptional machinery, significantly affecting the host immune response. This review explores the epigenetic regulation of autophagy during mycobacterium-host interactions. The interplay between epigenetic regulation and autophagy highlights a crucial aspect of host-pathogen interactions during Mtb infection. Understanding how Mtb manipulates the host epigenome to regulate autophagy could lead to the development of novel therapeutic strategies that enhance autophagic pathways or counteract Mtb's immune evasion tactics.
Collapse
Affiliation(s)
- Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Varsha Rawat
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| |
Collapse
|
4
|
Ost KJ, Student M, Cord-Landwehr S, Moerschbacher BM, Ram AFJ, Dirks-Hofmeister ME. Cell walls of filamentous fungi - challenges and opportunities for biotechnology. Appl Microbiol Biotechnol 2025; 109:125. [PMID: 40411627 PMCID: PMC12103488 DOI: 10.1007/s00253-025-13512-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/26/2025]
Abstract
The cell wall of filamentous fungi is essential for growth and development, both of which are crucial for fermentations that play a vital role in the bioeconomy. It typically has an inner rigid core composed of chitin and beta-1,3-/beta-1,6-glucans and a rather gel-like outer layer containing other polysaccharides and glycoproteins varying between and within species. Only a fraction of filamentous fungal species is used for the biotechnological production of enzymes, organic acids, and bioactive compounds such as antibiotics in large amounts on a yearly basis by precision fermentation. Most of these products are secreted into the production medium and must therefore pass through fungal cell walls at high transfer rates. Thus, cell wall mutants have gained interest for industrial enzyme production, although the causal relationship between cell walls and productivity requires further elucidation. Additionally, the extraction of valuable biopolymers like chitin and chitosan from spent fungal biomass, which is predominantly composed of cell walls, represents an underexplored opportunity for circular bioeconomy. Questions persist regarding the effective extraction of these biopolymers from the cell wall and their repurposing in valorization processes. This review aims to address these issues and promote further research on understanding the cell walls in filamentous fungi to optimize their biotechnological use. KEY POINTS: • The highly complex cell walls of filamentous fungi are important for biotechnology. • Cell wall mutants show promising potential to improve industrial enzyme secretion. • Recent studies revealed enhanced avenues for chitin/chitosan from fungal biomass.
Collapse
Affiliation(s)
- Katharina J Ost
- Laboratory for Food Biotechnology, Faculty of Agricultural Sciences and Landscape Architecture, Osnabrück University of Applied Sciences, Oldenburger Landstraße 62, 49090, Osnabrück, Germany
- Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143, Münster, Germany
| | - Mounashree Student
- Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143, Münster, Germany
| | - Stefan Cord-Landwehr
- Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143, Münster, Germany
| | - Bruno M Moerschbacher
- Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143, Münster, Germany
| | - Arthur F J Ram
- Fungal Genetics and Biotechnology, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE, Leiden, The Netherlands
| | - Mareike E Dirks-Hofmeister
- Laboratory for Food Biotechnology, Faculty of Agricultural Sciences and Landscape Architecture, Osnabrück University of Applied Sciences, Oldenburger Landstraße 62, 49090, Osnabrück, Germany.
| |
Collapse
|
5
|
Xu Y, Lee MK, de Weerd NA, Fu Z, Bertuzzo Veiga C, Dragoljevic D, Sviridov D, Hertzog PJ, Fleetwood AJ, Murphy AJ. Type I interferon signaling controls the early hematopoietic expansion in response to β-glucan. iScience 2025; 28:112347. [PMID: 40276764 PMCID: PMC12020881 DOI: 10.1016/j.isci.2025.112347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/02/2024] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Rapid hematopoietic adaptations are important for building and sustaining the biological response to β-glucan. The signals involved in these early events have not yet been fully explored. Given that type I interferons are produced in response to β-glucan and can profoundly impact hematopoietic stem cell (HSC) function, we hypothesized that this pathway may be involved in the early bone marrow response to β-glucan. In vivo administration of β-glucan led to local interferon-α production in the peritoneal cavity and bone marrow, upregulation of its receptor, IFNAR1, specifically on long-term hematopoietic stem cells (LT-HSCs), and broad expansion of downstream progenitor subpopulations. We demonstrate that intact type I interferon signaling is critical for β-glucan-mediated LT-HSC proliferation, mitochondrial activity, and glycolytic commitment. By determining that type I interferon signaling is important for LT-HSCs, which sit at the apex of the hematopoietic hierarchy, we uncover an important component of the early inflammatory response to β-glucan.
Collapse
Affiliation(s)
- Yangsong Xu
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Man K.S. Lee
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Nicole A. de Weerd
- Centre for Innate Immunity and Infectious Diseases, Department of Molecular and Translational Science, Hudson Institute of Medical Research and Monash University, Clayton, VIC, Australia
| | - Ziyue Fu
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Camilla Bertuzzo Veiga
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Dragana Dragoljevic
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Dmitri Sviridov
- Lipoproteins and Atherosclerosis, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Paul J. Hertzog
- Centre for Innate Immunity and Infectious Diseases, Department of Molecular and Translational Science, Hudson Institute of Medical Research and Monash University, Clayton, VIC, Australia
| | - Andrew J. Fleetwood
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Andrew J. Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| |
Collapse
|
6
|
Chaput S, Driouich JS, Gruber S, Busler D, de Lamballerie X, Nougairède A, Touret F. Assessing human liver spheroids as a model for antiviral drug evaluation against BSL-3 haemorrhagic fever viruses. Antiviral Res 2025; 239:106188. [PMID: 40360123 DOI: 10.1016/j.antiviral.2025.106188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 05/02/2025] [Accepted: 05/10/2025] [Indexed: 05/15/2025]
Abstract
Haemorrhagic fever viruses (HFVs) cause highly lethal syndromes with limited therapeutic options. Increasingly, 3D cell culture models are becoming an important tool in the field of virology. Since the liver is an important target for many HFVs, we evaluated a ready-to-use 96-well liver spheroid model composed of human primary cells for antiviral assessment. We worked with four biosafety level 3 (BSL-3) HFVs in this study: two orthoflaviviruses, Alkhumra haemorrhagic fever virus (AHFV) and yellow fever virus (YFV), and two viruses belonging to Hareavirales order, Pirital virus (PIRV), a surrogate for new-world BSL-4 mammarenaviruses, and Rift Valley fever virus (RVFV). We found that RVFV and PIRV were able to replicate in this model, whereas the orthoflaviviruses were not. A high viral dose was required for robust replication, and infectivity of RVFV in spheroids was low. We successfully demonstrated the antiviral activity of known broad-spectrum antiviral compounds-favipiravir, nitazoxanide, ribavirin, and galidesivir-despite some variability. However, except for ribavirin, higher doses were required in spheroids to detect antiviral effect compared to the 2D cell culture model. Overall, we conclude that human liver spheroids cannot replace traditional models for the selection of antiviral compounds but provide valuable additional complementary information. More broadly, this model could be useful to study viral pathogenicity and host-pathogen interactions.
Collapse
Affiliation(s)
- Sarah Chaput
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Jean-Sélim Driouich
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | | | | | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Antoine Nougairède
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Franck Touret
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France.
| |
Collapse
|
7
|
Li J, Wang H, Xia S. Hematopoietic stem and progenitor cells fine-tuning the "sweet" of trained immunity. J Leukoc Biol 2025; 117:qiaf043. [PMID: 40233187 DOI: 10.1093/jleuko/qiaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025] Open
Abstract
Recent studies have challenged the traditional view of innate immunity as nonspecific and transient by demonstrating that innate immune cells can develop immune memory in response to various activating factors, a phenomenon known as trained immunity. This process involves epigenetic modifications, such as changes in chromatin accessibility, and metabolic reprogramming, which can provide protection against unrelated pathogens but may also trigger immune-mediated damage. This review summarizes the current understanding of innate immune memory, with a particular focus on recent findings regarding the training of innate immune cells at the hematopoietic stem and progenitor cell stage. We present observations of trained immunity in innate immune cells, summarize key activating factors and underlying mechanisms, and propose potential host-directed immunotherapeutic strategies and preventive measures based on trained immunity. Our aim is to highlight the biological significance of trained immunity and its potential applications in enhancing long-term immunity, improving vaccine efficacy, and preventing immune-related diseases.
Collapse
Affiliation(s)
- Jiawei Li
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| | - Hui Wang
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| |
Collapse
|
8
|
Angulo M, Angulo C. Immunometabolic effects of β-glucan-trained immunity in newborn goats. Res Vet Sci 2025; 187:105612. [PMID: 40112655 DOI: 10.1016/j.rvsc.2025.105612] [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: 01/28/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Debaryomyces hansenii CBS 8339 β-glucans induced trained immunity in newborn goats. However, the metabolic shifts and potential signaling pathways have not been described yet. Thus, the present study aims to prove, firstly, modifications in cell metabolism related to trained immunity induction (β-glucans) and inhibition (MCC950) in an in vitro model upon lipopolysaccharide (LPS) re-stimulation; secondly, metabolic changes and possible signaling pathways are related to immune memory induced by β-glucan per os in newborns after ex vivo re-stimulation with a bacterial pathogen. Immune training leads to augmenting glycolysis (glucose and lactate) metabolites. Nevertheless, these changes were unaffected by a NOD-like receptor (NLRP3) inhibitor. In vivo training with oral β-glucan doses also evidenced an increase in glycolysis metabolites mediated by up-regulating AKT/MTOR/HIF1Α genes signaling pathway in monocytes; β-glucan in vivo training up-regulated Dectin1, TLR4, TLR6 RAF1, IL1Β and IL6 gene expressions in monocytes, while TNFΑ gene down-regulated. In conclusion, the results demonstrated that D. hansenii β-glucan induced trained immunity in newborn goat monocytes after LPS re-stimulation through glycolysis shifts, which were not reverted by the MCC950 inhibitor.
Collapse
Affiliation(s)
- Miriam Angulo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico, Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S. C.P. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico, Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S. C.P. 23096, Mexico.
| |
Collapse
|
9
|
Pellon A, Palacios A, Abecia L, Rodríguez H, Anguita J. Friends to remember: innate immune memory regulation by the microbiota. Trends Microbiol 2025; 33:510-520. [PMID: 39794207 DOI: 10.1016/j.tim.2024.12.002] [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: 10/18/2024] [Revised: 11/28/2024] [Accepted: 12/11/2024] [Indexed: 01/13/2025]
Abstract
Innate immune memory (IIM) is the process by which, upon a primary challenge, innate immune cells alter their epigenetic, transcriptional, and immunometabolic profiles, resulting in modified secondary responses. Unlike infections or other immune-system-related diseases, the role of IIM in nonpathogenic contexts is less understood. An increasing body of research has shown that normal microbiota members or their metabolic byproducts induce alternative memory phenotypes, suggesting that memory cells contribute to homeostasis in mucosal areas. In this review, we discuss the newest insights in the emerging field of IIM to the microbiota and the potential of manipulating these long-term responses to promote better mucosal health.
Collapse
Affiliation(s)
- Aize Pellon
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain.
| | - Ainhoa Palacios
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain; Present address: Research Unit, Basque Center for Blood Transfusion and Human Tissues, Osakidetza; Galdakao, Spain and Cell Therapy, Stem Cells and Tissues Group, BioBizkaia Health Research Institute; Barakaldo, Spain
| | - Leticia Abecia
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain; Department of Immunology, Microbiology, and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Héctor Rodríguez
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
10
|
Caronni N, La Terza F, Frosio L, Ostuni R. IL-1β + macrophages and the control of pathogenic inflammation in cancer. Trends Immunol 2025; 46:403-415. [PMID: 40169292 DOI: 10.1016/j.it.2025.03.001] [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: 01/07/2025] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 04/03/2025]
Abstract
While highlighting the complexity and heterogeneity of tumor immune microenvironments, the application of single-cell analyses in human cancers has identified recurrent subsets of tumor-associated macrophages (TAMs). Among these, interleukin (IL)-1β+ TAMs - cells with high levels of expression of inflammatory response and tissue repair genes, but with limited capacity to stimulate cytotoxic immunity - are emerging as key drivers of pathogenic inflammation in cancer. In this review we discuss recent literature defining the phenotypical, molecular, and functional properties of IL-1β+ TAMs, as well as their temporal dynamics and spatial organization. Elucidating the biology of these cells across tumor initiation, progression, metastasis, and therapy could inform the design and interpretation of clinical trials targeting IL-1β and/or other inflammatory factors in cancer immunotherapy.
Collapse
Affiliation(s)
- Nicoletta Caronni
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Federica La Terza
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Frosio
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Renato Ostuni
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
11
|
Chilibroste S, dos Santos JC, Mónaco A, Joosten LAB, Moreno M, Chabalgoity JA. Salmonella LVR01 triggers antagonistic two-armed innate immune memory that impacts on antitumor efficacy. Front Immunol 2025; 16:1535131. [PMID: 40370463 PMCID: PMC12075395 DOI: 10.3389/fimmu.2025.1535131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
The current understanding of innate immune memory encompasses both trained immunity and immune tolerance, where cells can exhibit enhanced responsiveness or immune paralysis upon subsequent stimuli, respectively. Various agents induce either of these responses, including β-glucan, Leishmania, BCG and LPS. BCG is a clinically approved immunotherapy for bladder cancer and BCG-induced trained immunity is important in driving anti-tumor adaptive immunity. Salmonella also shows promise in cancer treatment, eliciting potent anti-tumor immune responses, but with transitory effects. This led us to investigate whether Salmonella LVR01, like BCG, triggers trained immunity and its impact on anti-tumor responses. Herein, we report that Salmonella induces an enhanced response in bone marrow cells, characterized by a robust cytokine response upon a second stimulus, in a fashion that resembles trained immunity. Coherently with that, Salmonella administration induces enhanced responsiveness to a tumor implanted later in time, resulting in slow tumor growth and extended survival. However, in vitro stimulation of human monocytes and murine bone-marrow derived myeloid-enriched cells with Salmonella results in decreased production of cytokines resembling immune paralysis. Overall, our results suggest that Salmonella LVR01 induces enhanced responses of innate immune memory, as well as paralysis on monocytes. These two antagonistic effects could be the basis of the transitory effect of Salmonella treatment and suggest that further investigation on these phenomena could shed light on how to improve Salmonella-based immunotherapies for cancer.
Collapse
Affiliation(s)
- Sofía Chilibroste
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Jéssica C. dos Santos
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Amy Mónaco
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Leo A. B. Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - María Moreno
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - José A. Chabalgoity
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
12
|
Monreal-Escalante E, Angulo M, Ramos-Vega A, Trujillo E, Angulo C. Plant-made trained immunity-based vaccines: Beyond one approach. Int J Pharm 2025; 675:125572. [PMID: 40204041 DOI: 10.1016/j.ijpharm.2025.125572] [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: 02/06/2025] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Plant-made vaccines and trained immunity-based vaccines (TIbV or TRAIMbV) represent two strategies for enhancing immunity against diseases. Plants provide an effective and cost-efficient vaccine production platform, while TIbV induces innate immune memory that can protect against both homologous and heterologous diseases. Both strategies are generally compatible; however, they have not been explored in a transdisciplinary manner. Despite their strengths in vaccinology, each faces limitations that hinder widespread adoption and health benefits. This review revisits both strategies, discussing their fundamental knowledge alongside practical and experimental examples, ultimately highlighting their limitations and perspectives to pave the way for a unified approach to combat diseases. Future scenarios are envisioned and presented if research on plant-made trained immunity-based vaccines is adopted.
Collapse
Affiliation(s)
- Elizabeth Monreal-Escalante
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico; SECIHTI-Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico
| | - Miriam Angulo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico
| | - Abel Ramos-Vega
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Dirección: Boulevard de la Tecnología No.1036, Código Postal 62790 Xochitepec, Morelos, Mexico
| | - Edgar Trujillo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.
| |
Collapse
|
13
|
Ziogas A, Novakovic B, Ventriglia L, Galang N, Tran KA, Li W, Matzaraki V, van Unen N, Schlüter T, Ferreira AV, Moorlag SJCFM, Koeken VACM, Moyo M, Li X, Baltissen MPA, Martens JHA, Li Y, Divangahi M, Joosten LAB, Mhlanga MM, Netea MG. Long-term histone lactylation connects metabolic and epigenetic rewiring in innate immune memory. Cell 2025:S0092-8674(25)00400-3. [PMID: 40318634 DOI: 10.1016/j.cell.2025.03.048] [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: 04/26/2024] [Revised: 01/08/2025] [Accepted: 03/31/2025] [Indexed: 05/07/2025]
Abstract
Trained immunity, a de facto innate immune memory characterized by enhanced responsiveness to future challenges, is underpinned by epigenetic and metabolic rewiring. In individuals vaccinated with Bacille Calmette-Guérin (BCG), lactate release was associated with enhanced cytokine responsiveness upon restimulation. Trained monocytes/macrophages are characterized by lactylation of histone H3 at lysine residue 18(H3K18la), mainly at distal regulatory regions. Histone lactylation was positively associated with active chromatin and gene transcription, persisted after the elimination of the training stimulus, and was strongly associated with "trained" gene transcription in response to a secondary stimulus. Increased lactate production upon induction of trained immunity led to enhanced production of proinflammatory cytokines, a process associated with histone lactylation. Pharmacological inhibition of lactate production or histone lactylation blocked trained immunity responses, while polymorphisms of LDHA and EP300 genes modulated trained immunity. Long-term histone lactylation persisted in vivo 90 days after vaccination with BCG, highlighting H3K18la as an epigenetic mark of innate immune memory.
Collapse
Affiliation(s)
- Athanasios Ziogas
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands.
| | - Boris Novakovic
- Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Australia
| | - Lorenzo Ventriglia
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Hepatogastroenterology Division, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Noriko Galang
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Kim A Tran
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Wenchao Li
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Vasiliki Matzaraki
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nienke van Unen
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Titus Schlüter
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anaísa V Ferreira
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Simone J C F M Moorlag
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Valerie A C M Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Research Centre Innovations in Care, Rotterdam University of Applied Sciences, Rotterdam, the Netherlands
| | - Mthabisi Moyo
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Xiaolin Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marijke P A Baltissen
- Department of Molecular Biology, Faculty of Science, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Yang Li
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hanover Medical School, Hannover, Germany; Lower Saxony center for artificial intelligence and causal methods in medicine (CAIMed), Hannover, Germany
| | - Maziar Divangahi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Musa M Mhlanga
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.
| |
Collapse
|
14
|
Toner YC, Munitz J, Prevot G, Morla-Folch J, Wang W, van Elsas Y, Priem B, Deckers J, Anbergen T, Beldman TJ, Brechbühl EE, Aksu MD, Ziogas A, Sarlea SA, Ozturk M, Zhang Z, Li W, Li Y, Maier A, Fernandes JC, Cremers GA, van Genabeek B, Kreijtz JH, Lutgens E, Riksen NP, Janssen HM, Söntjens SH, Hoeben FJ, Kluza E, Singh G, Giamarellos-Bourboulis EJ, Schotsaert M, Duivenvoorden R, van der Meel R, Joosten LA, Cai L, Temel RE, Fayad ZA, Mhlanga MM, van Leent MM, Teunissen AJ, Netea MG, Mulder WJ. Targeting mTOR in myeloid cells prevents infection-associated inflammation. iScience 2025; 28:112163. [PMID: 40177636 PMCID: PMC11964677 DOI: 10.1016/j.isci.2025.112163] [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: 04/30/2024] [Revised: 12/13/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Infections, cancer, and trauma can cause life-threatening hyperinflammation. In the present study, using single-cell RNA sequencing of circulating immune cells, we found that the mammalian target of rapamycin (mTOR) pathway plays a critical role in myeloid cell regulation in COVID-19 patients. Previously, we developed an mTOR-inhibiting nanobiologic (mTORi-nanobiologic) that efficiently targets myeloid cells and their progenitors in the bone marrow. In vitro, we demonstrated that mTORi-nanobiologics potently inhibit infection-associated inflammation in human primary immune cells. Next, we investigated the in vivo effect of mTORi-nanobiologics in mouse models of hyperinflammation and acute respiratory distress syndrome. Using 18F-FDG uptake and flow cytometry readouts, we found mTORi-nanobiologic therapy to efficiently reduce hematopoietic organ metabolic activity and inflammation to levels comparable to those of healthy control animals. Together, we show that regulating myelopoiesis with mTORi-nanobiologics is a compelling therapeutic strategy to prevent deleterious organ inflammation in infection-related complications.
Collapse
Affiliation(s)
- Yohana C. Toner
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Jazz Munitz
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Geoffrey Prevot
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Judit Morla-Folch
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William Wang
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yuri van Elsas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Bram Priem
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Jeroen Deckers
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Tom Anbergen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Thijs J. Beldman
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Eliane E.S. Brechbühl
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Muhammed D. Aksu
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Athanasios Ziogas
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Sebastian A. Sarlea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Mumin Ozturk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Epigenomics & Single Cell Biophysics Group, Department of Cell Biology, FNWI, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Zhenhua Zhang
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
| | - Wenchao Li
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
| | - Alexander Maier
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cardiology and Angiology, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Jessica C. Fernandes
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Bas van Genabeek
- Trained Therapeutix Discovery, 5349 AB Oss, the Netherlands
- SyMO-Chem B.V., 5612 AZ Eindhoven, the Netherlands
| | | | - Esther Lutgens
- Department of Cardiovascular Medicine, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, MN 55905, USA
| | - Niels P. Riksen
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | | | | | | | - Ewelina Kluza
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Raphaël Duivenvoorden
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Nephrology, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, 400 349 Cluj-Napoca, Romania
| | - Lei Cai
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Ryan E. Temel
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Zahi A. Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Musa M. Mhlanga
- Epigenomics & Single Cell Biophysics Group, Department of Cell Biology, FNWI, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, 6525 GA Nijmegen, the Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Mandy M.T. van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abraham J.P. Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Willem J.M. Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
| |
Collapse
|
15
|
Sanchez Vasquez JD, Nkongolo S, Traum D, Sotov V, Kim SC, Mahamed D, Mehrotra A, Patel A, Chen DY, Fung S, Gaggar A, Feld JJ, Chang KM, Wallin JJ, Wang BX, Janssen HL, Gehring AJ. Virus-associated inflammation imprints an inflammatory profile on monocyte-derived macrophages in the human liver. J Clin Invest 2025; 135:e175241. [PMID: 40231469 PMCID: PMC11996867 DOI: 10.1172/jci175241] [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: 08/29/2023] [Accepted: 02/17/2025] [Indexed: 04/16/2025] Open
Abstract
Chronic liver injury triggers the activation and recruitment of immune cells, causing antigen-independent tissue damage and liver disease progression. Tissue inflammation can reshape macrophage composition through monocyte replacement. Replacement of tissue macrophages with monocytes differentiating in an inflammatory environment can potentially imprint a phenotype that switches the liver from an immune-tolerant organ to one predisposed to tissue damage. We longitudinally sampled the liver of patients with chronic hepatitis B who had active liver inflammation and were starting antiviral therapy. Antiviral therapy suppressed viral replication and liver inflammation, which coincided with decreased myeloid activation markers. Single-cell RNA-Seq mapped peripheral inflammatory markers to a monocyte-derived macrophage population, distinct from Kupffer cells, with an inflammatory transcriptional profile. The inflammatory macrophages (iMacs) differentiated from blood monocytes and were unique from macrophage found in healthy or cirrhotic liver. iMacs retained their core transcriptional signature after inflammation resolved, indicating inflammation-mediated remodeling of the macrophage population in the human liver that may affect progressive liver disease and immunotherapy.
Collapse
Affiliation(s)
- Juan Diego Sanchez Vasquez
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, and
- Schwartz Reisman Liver Research Centre, University Health Network, Toronto, Ontario, Canada
| | - Shirin Nkongolo
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Heidelberg, Germany
| | - Daniel Traum
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Medical Research, The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| | - Valentin Sotov
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | - Deeqa Mahamed
- Centre for Advanced Single Cell Analysis, The Hospital for Sick Children, Toronto, Canada
| | - Aman Mehrotra
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, and
| | | | | | - Scott Fung
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, and
| | - Anuj Gaggar
- Gilead Sciences, Foster City, California, USA
| | - Jordan J. Feld
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, and
| | - Kyong-Mi Chang
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Medical Research, The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| | | | - Ben X. Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Harry L.A. Janssen
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Adam J. Gehring
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, and
- Schwartz Reisman Liver Research Centre, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| |
Collapse
|
16
|
Xu C, Yang L, Cheng T, Wang Z, Liu C, Shao J. Sodium Houttuyfonate Ameliorates DSS-induced Colitis Aggravated by Candida albicans through Dectin-1/NF-κB/miR-32-5p/NFKBIZ Axis Based on Intestinal microRNA Profiling. Inflammation 2025; 48:820-838. [PMID: 38963571 DOI: 10.1007/s10753-024-02091-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Our previous research indicated that Sodium houttuyfonate (SH) can effectively ameliorate dextran sulfate sodium (DSS)-induced colitis exacerbated by Candida albicans. However, the underlying protective mechanism of SH remains unclear. Therefore, in this study, a mice colitis model was infected with C. albicans, and the total colonic miRNAs were assessed. Furthermore, the differentially expressed miRNAs were enriched, clustered, and analyzed. Moreover, based on the dual luciferase analysis of NFKBIZ modulation by miR-32-5p, the in vitro and in vivo therapeutic effects of SH on inflammatory response, fungal burden, oxidative stress, and apoptosis were assessed at transcriptional and translational levels in the presence of agonist and antagonist. A total of 1157 miRNAs were identified, 84 of which were differentially expressed. Furthermore, qRT-PCR validated that SH treatment improved 17 differentially expressed miRNAs with > fourfold upregulation or > sixfold downregulation. Similar to most differentially altered miRNA, C. albicans significantly increased Dectin-1, NF-κB, TNF-α, IL-1β, IL-17A, and decreased miR-32-5p which negatively targeted NFKBIZ. In addition, SH treatment reduced inflammatory response and fungal burden in a colitis model with C. albicans infection. Further analyses indicated that in C. albicans infected Caco2 cells, SH inhibited fungal growth, oxidative stress, and apoptosis by increasing Dectin-1, NF-κB, NFKBIZ, TNF-α, IL-1β, IL-17A, and decreasing miR-32-5p. Therefore, SH can ameliorate the severity of colitis aggravated by C. albicans via the Dectin-1/NF-κB/miR-32-5p/NFKBIZ axis.
Collapse
Affiliation(s)
- Chen Xu
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Liu Yang
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Ting Cheng
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Zixu Wang
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Chengcheng Liu
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Jing Shao
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China.
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China.
| |
Collapse
|
17
|
Minute L, Montalbán-Hernández K, Bravo-Robles L, Conejero L, Iborra S, Del Fresno C. Trained immunity-based mucosal immunotherapies for the prevention of respiratory infections. Trends Immunol 2025; 46:270-283. [PMID: 40113536 DOI: 10.1016/j.it.2025.02.012] [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: 01/08/2025] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
The devastating impact of respiratory infections demonstrates the critical need for novel prophylactic vaccines. In this opinion article, we advocate for bacterial immunotherapies as a complementary tool in our fight against respiratory infections. These immunotherapies can activate a wide spectrum of immunological mechanisms, with trained immunity (TI) being particularly significant. This phenomenon has led to the concept of trained immunity-based vaccines (TIbVs), which represent a novel approach in vaccinology. We discuss examples of TIbVs, including the tuberculosis vaccine Bacille Calmette-Guérin (BCG) and the polybacterial immunotherapy MV130. From our viewpoint, illustrating the mode of action and clinical evidence supports the proposal that TIbVs should be considered as next-generation vaccines to confer protection against a wide range of respiratory infections.
Collapse
Affiliation(s)
- Luna Minute
- The Innate Immune Response Group, La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain; Immunomodulation Laboratory, La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain
| | | | - Laura Bravo-Robles
- The Innate Immune Response Group, La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain; Immunomodulation Laboratory, La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain
| | | | | | - Carlos Del Fresno
- The Innate Immune Response Group, La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain; Immunomodulation Laboratory, La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain.
| |
Collapse
|
18
|
Simonis A, Theobald SJ, Koch AE, Mummadavarapu R, Mudler JM, Pouikli A, Göbel U, Acton R, Winter S, Albus A, Holzmann D, Albert MC, Hallek M, Walczak H, Ulas T, Koch M, Tessarz P, Hänsel-Hertsch R, Rybniker J. Persistent epigenetic memory of SARS-CoV-2 mRNA vaccination in monocyte-derived macrophages. Mol Syst Biol 2025; 21:341-360. [PMID: 40133533 PMCID: PMC11965535 DOI: 10.1038/s44320-025-00093-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Immune memory plays a critical role in the development of durable antimicrobial immune responses. How precisely mRNA vaccines train innate immune cells to shape protective host defense mechanisms remains unknown. Here we show that SARS-CoV-2 mRNA vaccination significantly establishes histone H3 lysine 27 acetylation (H3K27ac) at promoters of human monocyte-derived macrophages, suggesting epigenetic memory. However, we found that two consecutive vaccinations were required for the persistence of H3K27ac, which matched with pro-inflammatory innate immune-associated transcriptional changes and antigen-mediated cytokine secretion. H3K27ac at promoter regions were preserved for six months and a single mRNA booster vaccine potently restored their levels and release of macrophage-derived cytokines. Interestingly, we found that H3K27ac at promoters is enriched for G-quadruplex DNA secondary structure-forming sequences in macrophage-derived nucleosome-depleted regions, linking epigenetic memory to nucleic acid structure. Collectively, these findings reveal that mRNA vaccines induce a highly dynamic and persistent training of innate immune cells enabling a sustained pro-inflammatory immune response.
Collapse
Affiliation(s)
- Alexander Simonis
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Sebastian J Theobald
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Anna E Koch
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Ram Mummadavarapu
- Max Planck Research Group "Chromatin and Ageing", Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
| | - Julie M Mudler
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Andromachi Pouikli
- Max Planck Research Group "Chromatin and Ageing", Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
| | - Ulrike Göbel
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Richard Acton
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Babraham Institute, Cambridge, UK
| | - Sandra Winter
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Alexandra Albus
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Dmitriy Holzmann
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Marie-Christine Albert
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Institute of Biochemistry I, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Henning Walczak
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Institute of Biochemistry I, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, London, United Kingdom
| | - Thomas Ulas
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
- PRECISE Plattform for Single Cell Genomics and Epigenomics, DZNE, University of Bonn, Bonn and West German Genome Center, Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Manuel Koch
- Institute of Biochemistry I, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Dental, Oral and Maxillofacial Medicine (central facilities), Medical Faculty and University of Cologne, Cologne, Germany
| | - Peter Tessarz
- Max Planck Research Group "Chromatin and Ageing", Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department of Human Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Robert Hänsel-Hertsch
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute of Human Genetics, University Hospital Cologne, Cologne, Germany.
| | - Jan Rybniker
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany.
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany.
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
| |
Collapse
|
19
|
Almeida P, Alves I, Fernandes Â, Lima C, Freitas R, Braga I, Correia J, Jerónimo C, Pinho SS. "Mannose glycans as key players in trained immunity: A novel anti-tumoral catalyst". Biochim Biophys Acta Gen Subj 2025; 1869:130779. [PMID: 39988110 DOI: 10.1016/j.bbagen.2025.130779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
Cell wall glycans isolated from microorganisms are long known to provoke strong immune responses piloted by innate immune cell populations, including monocytes, in the context of Trained Immunity (TI). However, the contribution of yeast-derived mannan in the reprogramming of monocytes remains ill-defined. Here, we demonstrated that TI is often accompanied by an altered gene expression profile of selected glycan-binding proteins expressed by monocytes, including DC-SIGN and Dectin-2. Additionally, we showed that mannan, a mannose rich glycan, can trigger an enhanced immune phenotype compatible with TI in healthy monocytes, with glycan-primed cells exhibiting enhanced pro-inflammatory cytokine secretion (TNFα and IL-6) and higher activation (CD86) levels. Furthermore, the glycan-mediated priming of monocytes also imposed alterations to the expression of certain Glycan-Binding Proteins, such as DC-SIGN. Importantly, we established that these mannan-trained immune cells displayed an improved capacity to kill tumor cells in vitro. Lastly, we confirmed that monocytes from non-muscle invasive bladder cancer patients treated with BCG instillations presented a TI phenotype, as was revealed by the higher cytokine production and activation. Altogether, this study lays the foundations for exploiting the immunological potential of glycan-derived pathogens in reprogramming innate immune cells towards an effective anti-tumor immune response.
Collapse
Affiliation(s)
- Pedro Almeida
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal.
| | - Inês Alves
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal.
| | - Ângela Fernandes
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal.
| | - Cláudia Lima
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal.
| | - Rui Freitas
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal; Urology Clinic & Department of Urology, Portuguese Oncology Institute of Porto (IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal.
| | - Isaac Braga
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal; Urology Clinic & Department of Urology, Portuguese Oncology Institute of Porto (IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal.
| | - Jorge Correia
- Urology Clinic & Department of Urology, Portuguese Oncology Institute of Porto (IPOP), 4200-072 Porto, Portugal.
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal; Urology Clinic & Department of Urology, Portuguese Oncology Institute of Porto (IPOP), 4200-072 Porto, Portugal; RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal.
| | - Salomé S Pinho
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; ICBAS School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.
| |
Collapse
|
20
|
Jaiswal A, Halasz L, Williams DL, Osborne T. Setdb2 Regulates Inflammatory Trigger-Induced Trained Immunity of Macrophages Through Two Different Epigenetic Mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.644013. [PMID: 40166182 PMCID: PMC11956931 DOI: 10.1101/2025.03.18.644013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
"Trained immunity" of innate immune cells occurs through a sequential two-step process where an initial pathogenic or sterile inflammatory trigger is followed by an amplified response to a later un-related secondary pathogen challenge. The memory effect is mediated at least in part through epigenetic modifications of the chromatin landscape. Here, we investigated the role of the epigenetic modifier Setdb2 in microbial (β-glucan) or sterile trigger (Western-diet-WD/oxidized-LDL-oxLDL)-induced trained immunity of macrophages. Using genetic mouse models and genomic analysis, we uncovered a critical role of Setdb2 in regulating proinflammatory and metabolic pathway reprogramming. We further show that Setdb2 regulates trained immunity through two different complementary mechanisms: one where it positively regulates glycolytic and inflammatory pathway genes via enhancer-promoter looping, and is independent of its enzymatic activity; while the second mechanism is associated with both increased promoter associated H3K9 methylation and repression of interferon response pathway genes. Interestingly, while both mechanisms occur in response to pathogenic training, only the chromatin-looping mechanism operates in response to the sterile inflammatory stimulus. These results reveal a previously unknown bifurcation in the downstream pathways that distinguishes between pathogenic and sterile inflammatory signaling responses associated with the innate immune memory response and may provide potential therapeutic opportunities to target cytokine vs. interferon pathways to limit complications of chronic inflammation.
Collapse
|
21
|
Rehill AM, McCluskey S, Ledwith AE, Ryan TAJ, Ünlü B, Leon G, Charles-Messance H, Gilbert EH, Klavina P, Day EA, Coppinger J, O’Sullivan JM, McMahon C, O’Donnell JS, Curtis AM, O’Neill LAJ, Sheedy FJ, Preston RJS. Trained immunity causes myeloid cell hypercoagulability. SCIENCE ADVANCES 2025; 11:eads0105. [PMID: 40053582 PMCID: PMC11887800 DOI: 10.1126/sciadv.ads0105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
The pathogenic basis for increased thrombotic risk in individuals with inflammatory diseases is poorly understood. Myeloid cell "trained immunity" describes persistent innate immune cell memory arising from prior exposure to an inflammatory stimulus, leading to an enhanced immune response to subsequent unrelated stimuli. We identify enhanced myeloid cell prothrombotic activity as a maladaptive consequence of trained immunity. Lipopolysaccharide (LPS) stimulation of macrophages trained previously with β-glucan or heme exhibited significantly enhanced procoagulant activity compared to macrophages stimulated with LPS alone, which was mediated by enhanced acid sphingomyelinase-mediated tissue factor decryption. Furthermore, splenic monocytes isolated from β-glucan-trained mice revealed enhanced procoagulant activity up to 4 weeks after β-glucan administration compared to monocytes from control mice over the same time period. Moreover, hematopoietic progenitor cells and bone marrow interstitial fluid from β-glucan-trained mice had enhanced procoagulant activity compared to control mice. Trained immunity and associated metabolic perturbations may therefore represent an opportunity for targeted intervention in immunothrombotic disease development.
Collapse
Affiliation(s)
- Aisling M. Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | - Seán McCluskey
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | - Anna E. Ledwith
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Tristram A. J. Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Betül Ünlü
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | | | - Edmund H. Gilbert
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paula Klavina
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | - Emily A. Day
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Judith Coppinger
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Jamie M. O’Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Corrina McMahon
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | - James S. O’Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Annie M. Curtis
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Luke A. J. O’Neill
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Frederick J. Sheedy
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Roger J. S. Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| |
Collapse
|
22
|
Martín‐Cruz L, Palomares O. Allergen-Specific Immunotherapy and Trained Immunity. Allergy 2025; 80:677-689. [PMID: 39641571 PMCID: PMC11891420 DOI: 10.1111/all.16423] [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: 08/08/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
The high prevalence of allergic diseases reached over the last years is attributed to the complex interplay of genetic factors, lifestyle changes, and environmental exposome. Allergen-specific immunotherapy (AIT) is the single therapeutic strategy for allergic diseases with the potential capacity to modify the course of the disease. Our knowledge of the mechanisms involved in allergy and successful AIT has significantly improved. Recent findings indicate that long-term allergen tolerance upon AIT discontinuation not only relies on the generation of proper adaptive immune responses by the generation of allergen-specific regulatory T and B cells enabling the induction of different isotypes of blocking antibodies but also relies on the restoration of proper innate immune responses. Trained immunity (TRIM) is the process by which innate immune cells acquire memory by mechanisms depending on metabolic and epigenetic reprogramming, thus conferring the host with increased broad protection against infection. This concept was initially explored for infectious diseases, as well as for vaccination against infections, but compelling experimental evidence suggests that TRIM might also play a role in allergy and AIT. Hyperinflammatory innate immune responses in early life, likely due to TRIM maladaptations, lead to aberrant type 2 inflammation-enhancing allergy. However, exposure to farming environments and specific microbes prevents recurrent infections and allergy development, likely due to mechanisms partially depending on TRIM. TRIM-based vaccines and next-generation AIT vaccines inducing metabolic and epigenetic reprogramming in innate immune cells and their precursors have shown protective antiallergic effects. A better understanding of the factors involved in early-life TRIM mechanisms in the context of allergy and the identification and characterization of novel tolerance inducers might well enable the design of alternative TRIM-based allergen vaccines for allergic diseases.
Collapse
Affiliation(s)
- Leticia Martín‐Cruz
- School of Chemistry, Department of Biochemistry and Molecular BiologyComplutense UniversityMadridSpain
- School of Pharmacy, Department of Biochemistry and Molecular BiologyComplutense UniversityMadridSpain
| | - Oscar Palomares
- School of Chemistry, Department of Biochemistry and Molecular BiologyComplutense UniversityMadridSpain
| |
Collapse
|
23
|
Li Y, Wang L, Chen S. An overview of the progress made in research into the Mpox virus. Med Res Rev 2025; 45:788-812. [PMID: 39318037 DOI: 10.1002/med.22085] [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: 03/26/2024] [Revised: 08/05/2024] [Accepted: 09/01/2024] [Indexed: 09/26/2024]
Abstract
Mpox is a zoonotic illness caused by the Mpox virus (MPXV), a member of the Orthopoxvirus family. Although a few cases have been reported outside Africa, it was originally regarded as an endemic disease limited to African countries. However, the Mpox outbreak of 2022 was remarkable in that the infection spread to more than 123 countries worldwide, causing thousands of infections and deaths. The ongoing Mpox outbreak has been declared as a public health emergency of international concern by the World Health Organization. For a better management and control of the epidemic, this review summarizes the research advances and important scientific findings on MPXV by reviewing the current literature on epidemiology, clinical characteristics, diagnostic methods, prevention and treatment measures, and animal models of MPXV. This review provides useful information to raise awareness about the transmission, symptoms, and protective measures of MPXV, serving as a theoretical guide for relevant institutions to control MPXV.
Collapse
Affiliation(s)
- Yansheng Li
- Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound lmaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Department of Critical Care Medicine, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Lianrong Wang
- Department of Respiratory Diseases, Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Shi Chen
- Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound lmaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Department of Critical Care Medicine, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
24
|
Zhao Y, Zhou Z, Cai G, Zhang D, Yu X, Li D, Li S, Zhang Z, Zhang D, Luo J, Hu Y, Gao A, Zhang H. Systemic infection by Candida albicans requires FASN-α subunit induced cell wall remodeling to perturb immune response. PLoS Pathog 2025; 21:e1012865. [PMID: 40138332 PMCID: PMC11940687 DOI: 10.1371/journal.ppat.1012865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/29/2024] [Indexed: 03/29/2025] Open
Abstract
Invasive fungal infections are a leading cause of mortality and morbidity in patients with severely impaired host defenses, while treatment options remain limited. Fatty acid synthase (FASN), the key enzyme regulating de novo biosynthesis of fatty acids, is crucial for the lethal infection of fungi; however, its pathogenic mechanism is still far from clear. Here, we identified the α subunit of FASN as a potential immunotherapeutic target against systemic Candida albicans infection. The avirulence of the encoded gene (FAS2) -deleted mutant in a mouse model of systemic candidiasis is not due to its fitness defects, because sufficient exogenous fatty acids in serum can overcome FASN inhibition. However, the FAS2-deleted mutant displays increased circulating innate immune responses and enhances activated neutrophil fungicidal activity through the unmasking of immunogenic cell wall epitopes via the Rho-1 dependent Mkc1-MAPK signaling pathway, which facilitates fungal clearance, reduces renal tissue damage and inflammatory cell infiltration, ultimately lowers fungal pathogenicity. Priming with the FAS2-deleted mutant provided significant protection against subsequent lethal infection with wild-type C. albicans in mice as early as one week, and it was well-tolerated with limited toxicity. Our findings indicate that the FASN-α subunit plays key roles in the regulation of neutrophil-associated antifungal immunity and could be a potential target for immunotherapeutic intervention.
Collapse
Affiliation(s)
- Yajing Zhao
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Zhishan Zhou
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Guiyue Cai
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Dandan Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Xiaoting Yu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington District of Columbia, United States of America
| | - Shuixiu Li
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Zhanpeng Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Dongli Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Jiyao Luo
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| | - Yunfeng Hu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Aili Gao
- Guangzhou Dermatology Hospital, Guangzhou, China
| | - Hong Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Mycology, Jinan University, Guangzhou, China
| |
Collapse
|
25
|
Askary A, Chen W, Choi J, Du LY, Elowitz MB, Gagnon JA, Schier AF, Seidel S, Shendure J, Stadler T, Tran M. The lives of cells, recorded. Nat Rev Genet 2025; 26:203-222. [PMID: 39587306 DOI: 10.1038/s41576-024-00788-w] [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] [Accepted: 09/26/2024] [Indexed: 11/27/2024]
Abstract
A paradigm for biology is emerging in which cells can be genetically programmed to write their histories into their own genomes. These records can subsequently be read, and the cellular histories reconstructed, which for each cell could include a record of its lineage relationships, extrinsic influences, internal states and physical locations, over time. DNA recording has the potential to transform the way that we study developmental and disease processes. Recent advances in genome engineering are driving the development of systems for DNA recording, and meanwhile single-cell and spatial omics technologies increasingly enable the recovery of the recorded information. Combined with advances in computational and phylogenetic inference algorithms, the DNA recording paradigm is beginning to bear fruit. In this Perspective, we explore the rationale and technical basis of DNA recording, what aspects of cellular biology might be recorded and how, and the types of discovery that we anticipate this paradigm will enable.
Collapse
Affiliation(s)
- Amjad Askary
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Wei Chen
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Junhong Choi
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lucia Y Du
- Biozentrum, University of Basel, Basel, Switzerland
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Michael B Elowitz
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA, USA.
| | - James A Gagnon
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA.
| | - Alexander F Schier
- Biozentrum, University of Basel, Basel, Switzerland.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| | - Sophie Seidel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
| | - Tanja Stadler
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Martin Tran
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
26
|
Qi R, Cheng X, Chen S, Fan J. Extracellular HSP70 facilitated β-glucan induced trained immunity in macrophages to suppress sepsis via TLR2-NF-κB axis. Cytokine 2025; 187:156861. [PMID: 39823994 DOI: 10.1016/j.cyto.2025.156861] [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: 12/01/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/20/2025]
Abstract
Sepsis is a common systemic infectious disease followed by extremely high incidence and mortality with no effective treatment and clinical drugs. As a key mediator involved in infection and immunity, it has been reported that sepsis patients are accompanied by increased heat shock protein 70 (HSP70). Trained immunity is a novel innate immunity approach that can be activated by β-glucan to fight against sepsis. The mechanism of HSP70 activating trained macrophages against sepsis needs further elucidation. Trained immunity and sepsis models were established by β-glucan and LPS individually both in vivo and in vitro. We demonstrated that HSP70 was significantly upregulated in septic mice serum, and HSP70 could protect mice from sepsis by activating β-glucan-trained macrophages as an ideal secondary inducer via TLR2-NF-κB pathway. Additionally, the sepsis resistant effects of HSP70 could be blocked by its antibody. In summary, more than a molecular chaperone to maintain homeostasis, HSP70 could be an important trained immunity inducer to help the body fighting against sepsis, which provided new stimuli for trained immunity and novel therapeutic solutions for sepsis.
Collapse
Affiliation(s)
- Ran Qi
- Department of Clinical Laboratory, The Second Children & Women's Healthcare of Jinan City, Jinan, Shandong, China
| | - Xin Cheng
- Department of Clinical Laboratory, Jinan City People's Hospital, Jinan, Shandong, China
| | - Shan Chen
- Department of Clinical Laboratory, The Second Children & Women's Healthcare of Jinan City, Jinan, Shandong, China
| | - Jinjun Fan
- Department of Clinical Laboratory, The Second Children & Women's Healthcare of Jinan City, Jinan, Shandong, China.
| |
Collapse
|
27
|
Hu W, Shi S, Lin J, Gao T, Shen J, Sun Y, Wei H, Zheng X. The lower rate of bone and joint infection in patients with open extremity fractures associated with vaccination prior to injury: a propensity-matched cohort study. Front Pharmacol 2025; 16:1546191. [PMID: 40051567 PMCID: PMC11882553 DOI: 10.3389/fphar.2025.1546191] [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: 12/16/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Background Vaccines could strengthen the innate immune system in addition to conferring protection against their target pathogen via vaccine-induced immunomodulation, a phenomenon termed trained immunity. The purpose of the present study was to determine whether vaccination prior to injury is associated with a lower rate of bone and joint infections (BJIs) in patients with open extremity fractures. Methods Patients with open extremity fractures treated at one hospital between January 2010 and December 2019 were identified. Incidental vaccine recipients and control cohorts were matched in a 1:1 ratio using propensity scores based on age, sex, anatomical location of the fracture, Gustilo-Anderson classification, body mass index (BMI), and diagnosis of diabetes. The primary endpoint was BJIs within 1 year after initial injury. Secondary outcomes were neutrophil counts and serum C-reactive protein (CRP) levels within 24 h of admission. Logistic or linear regression was performed to control for potential confounding factors when comparing primary and secondary outcomes. Results Vaccine inoculation history was successfully collected from 6,338 patients, with only 83 patients receiving an incidental vaccine inoculation within 3 months before injury. After propensity score matching, demographic and clinical factors were well-balanced between cohorts (all standardized differences >0.1). After controlling for potential confounders, patients in the vaccine group were at a lower risk of BJIs after open extremity fractures (vaccine, 2/83 [2.4%]; control, 10/83 [12.0%), p = 0.011). Levels of circulating neutrophils and CRP were slightly increased in the vaccine group. Conclusion Vaccine inoculation is associated with the lower BJI rate after open extremity fractures, and vaccinated patients might have a more robust immune response against bacterial challenges in terms of neutrophil and CRP levels after injury. Future prospective cohort studies and clinical trials are warranted to evaluate this finding definitively. Clinical Trail registration http://www.chictr.org.cn/usercenter.aspx, identifier ChiCTR2000041093.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xianyou Zheng
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Haacke N, Wang H, Yan S, Barovic M, Li X, Nagai K, Botezatu A, Hatzioannou A, Gercken B, Trimaglio G, Shah AU, Wang J, Ye L, Jaykar MT, Rauner M, Wielockx B, Chung KJ, Netea MG, Kalafati L, Hajishengallis G, Chavakis T. Innate immune training of osteoclastogenesis promotes inflammatory bone loss in mice. Dev Cell 2025:S1534-5807(25)00063-2. [PMID: 40020679 PMCID: PMC7617534 DOI: 10.1016/j.devcel.2025.02.001] [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: 05/07/2024] [Revised: 12/06/2024] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
We previously demonstrated that long-term trained immunity (TRIM) involves adaptations that imprint innate immune memory in long-lived myelopoiesis precursors and their progeny, monocytes/macrophages and neutrophils, which thereby acquire enhanced responsiveness to future challenges. Here, we show that a distinct component of myeloid biology, osteoclastogenesis, can also undergo innate immune training. Indeed, β-glucan-induced TRIM was associated with an increased osteoclastogenesis bias in the bone marrow and an expansion of monocytes/osteoclast progenitors in the periphery, resulting in aggravated severity of experimental periodontitis and arthritis. In the setting of trained inflammatory osteoclastogenesis, we observed transcriptomic rewiring in synovial myeloid cells of arthritic mice, featuring prominent upregulation of the transcription factor melanogenesis-associated transcription factor (MITF). Adoptive transfer of splenic monocytes from β-glucan-trained mice to naive recipients exacerbated arthritis in the latter in a strictly MITF-dependent manner. Our findings establish trained osteoclastogenesis as a maladaptive component of TRIM and potentially provide therapeutic targets in inflammatory bone loss disorders.
Collapse
Affiliation(s)
- Nora Haacke
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Hui Wang
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shu Yan
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany
| | - Marko Barovic
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Xiaofei Li
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kosuke Nagai
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Adelina Botezatu
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Aikaterini Hatzioannou
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Giulia Trimaglio
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany
| | - Anisha U Shah
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mangesh T Jaykar
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 XZ Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Lydia Kalafati
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| |
Collapse
|
29
|
Wang AG, Son M, Gorin A, Kenna E, Padhi A, Keisham B, Schauer A, Hoffmann A, Tay S. Macrophage memory emerges from coordinated transcription factor and chromatin dynamics. Cell Syst 2025; 16:101171. [PMID: 39938520 DOI: 10.1016/j.cels.2025.101171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/18/2024] [Accepted: 01/09/2025] [Indexed: 02/14/2025]
Abstract
Cells of the immune system operate in dynamic microenvironments where the timing, concentration, and order of signaling molecules constantly change. Despite this complexity, immune cells manage to communicate accurately and control inflammation and infection. It is unclear how these dynamic signals are encoded and decoded and if individual cells retain the memory of past exposure to inflammatory molecules. Here, we use live-cell analysis, ATAC sequencing, and an in vivo model of sepsis to show that sequential inflammatory signals induce memory in individual macrophages through reprogramming the nuclear factor κB (NF-κB) network and the chromatin accessibility landscape. We use transcriptomic profiling and deep learning to show that transcription factor and chromatin dynamics coordinate fine-tuned macrophage responses to new inflammatory signals. This work demonstrates how macrophages retain the memory of previous signals despite single-cell variability and elucidates the mechanisms of signal-induced memory in dynamic inflammatory conditions like sepsis.
Collapse
Affiliation(s)
- Andrew G Wang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Medical Scientist Training Program, University of Chicago, Chicago, IL 60637, USA
| | - Minjun Son
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Chan Zuckerberg Biohub Chicago, Chicago, IL, USA
| | - Aleksandr Gorin
- Department of Medicine, Division of Infectious Diseases, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Emma Kenna
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Abinash Padhi
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Bijentimala Keisham
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Adam Schauer
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA
| | - Alexander Hoffmann
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Savaş Tay
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
30
|
Jurczak M, Druszczynska M. Beyond Tuberculosis: The Surprising Immunological Benefits of the Bacillus Calmette-Guérin (BCG) Vaccine in Infectious, Auto-Immune, and Inflammatory Diseases. Pathogens 2025; 14:196. [PMID: 40005571 PMCID: PMC11857995 DOI: 10.3390/pathogens14020196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/08/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The Bacillus Calmette-Guérin (BCG) vaccine, best known for its role in preventing tuberculosis, has recently garnered attention for its broader immunomodulatory effects. By inducing trained immunity, BCG reprograms innate immune cells, enhancing their responses to various pathogens. This process, driven by epigenetic and metabolic reprogramming, suggests that BCG may have therapeutic potential far beyond tuberculosis. Emerging evidence points to its potential benefits in conditions such as autoimmune diseases, cancer, and viral infections. Furthermore, by modulating immune activity, BCG has been proposed to reduce chronic inflammation and promote immune tolerance. This review delves into the multifaceted role of BCG, highlighting its potential as a versatile therapeutic tool for managing a wide range of diseases.
Collapse
Affiliation(s)
- Magdalena Jurczak
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, Institute of Microbiology, Biotechnology and Immunology, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland;
- The Bio-Med-Chem Doctoral of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, 90-237 Lodz, Poland
- Department of Microbiology and Experimental Immunology, MOLecoLAB: Lodz Centre of Molecular Studies on Civilisation Diseases, Medical University of Lodz, 92-215 Lodz, Poland
| | - Magdalena Druszczynska
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, Institute of Microbiology, Biotechnology and Immunology, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland;
| |
Collapse
|
31
|
Robles-Vera I, Jarit-Cabanillas A, Brandi P, Martínez-López M, Martínez-Cano S, Rodrigo-Tapias M, Femenía-Muiña M, Redondo-Urzainqui A, Nuñez V, González-Correa C, Moleón J, Duarte J, Conejero L, Mata-Martínez P, Díez-Rivero CM, Bergón-Gutiérrez M, Fernández-López I, Gómez MJ, Quintas A, Dopazo A, Sánchez-Cabo F, Pariente E, Del Fresno C, Subiza JL, Iborra S, Sancho D. Microbiota translocation following intestinal barrier disruption promotes Mincle-mediated training of myeloid progenitors in the bone marrow. Immunity 2025; 58:381-396.e9. [PMID: 39848243 PMCID: PMC11832192 DOI: 10.1016/j.immuni.2024.12.012] [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: 08/02/2024] [Revised: 10/29/2024] [Accepted: 12/31/2024] [Indexed: 01/25/2025]
Abstract
Impairment of the intestinal barrier allows the systemic translocation of commensal bacteria, inducing a proinflammatory state in the host. Here, we investigated innate immune responses following increased gut permeability upon administration of dextran sulfate sodium (DSS) in mice. We found that Enterococcus faecalis translocated to the bone marrow following DSS treatment and induced trained immunity (TI) hallmarks in bone-marrow-derived mouse macrophages and human monocytes. DSS treatment or heat-killed E. faecalis reprogrammed bone marrow progenitors (BMPs), resulting in enhanced inflammatory responses in vitro and in vivo and protection against subsequent pathogen infections. The C-type lectin receptor Mincle (Clec4e) was essential for E. faecalis-induced TI in BMPs. Clec4e-/- mice showed impaired TI upon E. faecalis administration and reduced pathology following DSS treatment. Thus, Mincle sensing of E. faecalis induces TI that may have long-term effects on pathologies associated with increased gut permeability.
Collapse
Affiliation(s)
- Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| | - Aitor Jarit-Cabanillas
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Paola Brandi
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Sarai Martínez-Cano
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Inmunotek S.L., Alcalá de Henares, Spain
| | | | | | | | - Vanesa Nuñez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cristina González-Correa
- Department of Pharmacology, School of Pharmacy, University of Granada, IBS-Granada, Centro de Investigaciones Biomédicas (CIBM), CIBER-Enfermedades Cardiovasculares (CiberCV), Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy, University of Granada, IBS-Granada, Centro de Investigaciones Biomédicas (CIBM), CIBER-Enfermedades Cardiovasculares (CiberCV), Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy, University of Granada, IBS-Granada, Centro de Investigaciones Biomédicas (CIBM), CIBER-Enfermedades Cardiovasculares (CiberCV), Granada, Spain
| | | | - Pablo Mata-Martínez
- Immunomodulation Lab, Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | | | - Marta Bergón-Gutiérrez
- Immunomodulation Lab, Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | | | - Manuel J Gómez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Quintas
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Esther Pariente
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Carlos Del Fresno
- Immunomodulation Lab, Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | | | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain; Fundación Inmunotek, Alcalá de Henares, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
32
|
Doyle RM, Kannambath S, Pittman A, Goliath R, Kumar V, Meintjes G, Milburn J, Netea MG, Harrison TS, Jarvis JN, Bicanic T. Ex Vivo Host Transcriptomics During Cryptococcus neoformans, Cryptococcus gattii, and Candida albicans Infection of Peripheral Blood Mononuclear Cells From South African Volunteers. J Infect Dis 2025; 231:e254-e262. [PMID: 39158391 PMCID: PMC11793030 DOI: 10.1093/infdis/jiae410] [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: 04/06/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024] Open
Abstract
Cryptococcus neoformans, Cryptococcus gattii, and Candida albicans are opportunistic fungal pathogens associated with infections in immunocompromised hosts. Cryptococcal meningitis (CM) is the leading fungal cause of human immunodeficiency virus-related deaths globally, with the majority occurring in Africa. The human immune response to C albicans infection has been studied extensively in large genomics studies whereas cryptococcal infections, despite their severity, are comparatively understudied. Here we investigated the transcriptional response of immune cells after in vitro stimulation with in vitro C neoformans, C gattii, and C albicans infection of peripheral blood mononuclear cells collected from healthy South African volunteers. We found a lower transcriptional response to cryptococcal stimuli compared to C albicans and unique expression signatures from all 3 fungal stimuli. This work provides a starting point for further studies comparing the transcriptional signature of CM in immunocompromised patients, with the goal of identifying biomarkers of disease severity and possible novel treatment targets.
Collapse
Affiliation(s)
- Ronan M Doyle
- Department of Clinical Research, London School of Hygiene and Tropical Medicine
| | - Shichina Kannambath
- Genomics Facility, The Institute of Cancer Research
- Institute of Infection and Immunity, St George's University London, and Clinical Academic Group in Infection, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Alan Pittman
- Institute of Infection and Immunity, St George's University London, and Clinical Academic Group in Infection, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Rene Goliath
- Department of Medicine and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - Vinod Kumar
- Botswana Harvard Health Partnership, Gaborone
| | - Graeme Meintjes
- Department of Medicine and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - James Milburn
- Department of Clinical Research, London School of Hygiene and Tropical Medicine
- Botswana Harvard Health Partnership, Gaborone
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas S Harrison
- Institute of Infection and Immunity, St George's University London, and Clinical Academic Group in Infection, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
- Medical Research Council Centre for Medical Mycology, University of Exeter, United Kingdom
| | - Joseph N Jarvis
- Department of Clinical Research, London School of Hygiene and Tropical Medicine
- Botswana Harvard Health Partnership, Gaborone
| | - Tihana Bicanic
- Institute of Infection and Immunity, St George's University London, and Clinical Academic Group in Infection, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
33
|
Hoseinzadeh A, Esmaeili SA, Sahebi R, Melak AM, Mahmoudi M, Hasannia M, Baharlou R. Fate and long-lasting therapeutic effects of mesenchymal stromal/stem-like cells: mechanistic insights. Stem Cell Res Ther 2025; 16:33. [PMID: 39901306 PMCID: PMC11792531 DOI: 10.1186/s13287-025-04158-z] [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: 04/27/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
A large body of evidence suggests that mesenchymal stromal cells (MSCs) are able to respond rapidly to the cytokine milieu following systemic infusion. This encounter has the potential to dictate their therapeutic efficacy (also referred to as licensing). MSCs are able to rapidly react to cellular damage by migrating to the inflamed tissue and ultimately modifying the inflammatory microenvironment. However, the limited use of MSCs in clinical practice can be attributed to a lack of understanding of the fate of MSCs in patients after administration and long term MSC-derived therapeutic activity. While the known physiological effectors of viable MSCs make a relative contribution, an innate property of MSCs as a therapeutic agent is their caspase-dependent cell death. These mechanisms may be involving the functional reprogramming of myeloid phagocytes via efferocytosis, the process by which apoptotic bodies (ABs) are identified for engulfment by both specialized and non-specialized phagocytic cells. Recent studies have provided evidence that the uptake of ABs with a distinct genetic component can induce changes in gene expression through the process of epigenetic remodeling. This phenomenon, known as 'trained immunity', has a significant impact on immunometabolism processes. It is hypothesized that the diversity of recipient cells within the inflammatory stroma adjacent to MSCs may potentially serve as a biomarker for predicting the clinical outcome of MSC treatment, while also contributing to the variable outcomes observed with MSC-based therapies. Therefore, the long-term reconstructive process of MSCs may potentially be mediated by MSC apoptosis and subsequent phagocyte-mediated efferocytosis.
Collapse
Affiliation(s)
- Akram Hoseinzadeh
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed-Alireza Esmaeili
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Sahebi
- Department of Modern Sciences and Technologies, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mahmoud Mahmoudi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maliheh Hasannia
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Rasoul Baharlou
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
34
|
Luo Y, Jiang LY, Liao ZZ, Wang YY, Wang YD, Xiao XH. Metabolic Regulation of Inflammation: Exploring the Potential Benefits of Itaconate in Autoimmune Disorders. Immunology 2025; 174:189-202. [PMID: 39542834 DOI: 10.1111/imm.13875] [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: 09/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
Itaconic acid and its metabolites have demonstrated significant therapeutic potential in various immune diseases. Originating from the tricarboxylic acid cycle in immune cells, itaconic acid can modulate immune responses, diminish inflammation, and combat oxidative stress. Recent research has uncovered multiple mechanisms through which itaconic acid exerts its effects, including the inhibition of inflammatory cytokine production, activation of anti-inflammatory pathways, and modulation of immune cell function by regulating cellular metabolism. Cellular actions are influenced by the modulation of metabolic pathways, such as inhibiting succinate dehydrogenase (SDH) activity or glycolysis, activation of nuclear-factor-E2-related factor 2 (Nrf2), boosting cellular defences against oxidative stress, and suppression of immune cell inflammation through the NF-κB pathway. This comprehensive review discusses the initiation, progression, and mechanisms of action of itaconic acid and its metabolites, highlighting their modulatory effects on various immune cell types. Additionally, it examines their involvement in immune disease like rheumatoid arthritis, multiple sclerosis, type 1 diabetes mellitus, and autoimmune hepatitis, offering greater understanding for creating new therapies for these ailments.
Collapse
Affiliation(s)
- Yin Luo
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li-Yan Jiang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhe-Zhen Liao
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
35
|
Wang C, De Francesco R, Lamers LA, Rinzema S, Frölich S, van Lent PLEM, Logie C, van den Bosch MHJ. Transcriptomic profiling of osteoarthritis synovial macrophages reveals a tolerized phenotype compounded by a weak corticosteroid response. Rheumatology (Oxford) 2025; 64:860-869. [PMID: 38466933 PMCID: PMC11781583 DOI: 10.1093/rheumatology/keae161] [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: 04/12/2023] [Revised: 01/09/2024] [Accepted: 03/01/2024] [Indexed: 03/13/2024] Open
Abstract
OBJECTIVES It is well-known that long-term osteoarthritis prognosis is not improved by corticosteroid treatments. Here we investigate what could underlie this phenomenon by measuring the short term corticosteroid response of osteoarthritic joint synovial macrophages (OA-Mf). METHODS We determined the genome-wide transcriptomic response to corticosteroids of end-stage OA-Mf. This was compared with lipopolysaccharide-tolerized and β-glucan-trained circulating blood monocyte-derived macrophage models. RESULTS Upon corticosteroid stimulation, the trained and tolerized macrophages significantly altered the abundance of 201 and 257 RNA transcripts, respectively. By contrast, by the same criteria, OA-Mf had a very restricted corticosteroid response of only 12 RNA transcripts. Furthermore, while metalloproteinases 1, 2, 3 and 10 expression clearly distinguish OA-Mf from both the tolerized and trained macrophage models, OA-Mf IL-1, chemokine (CXCL) and cytokine (CCL) family member profiles resembled the tolerized macrophage model, with the exception that OA-Mf showed high levels of CCL20. CONCLUSION Terminal osteoarthritis joints harbour macrophages with an inflammatory state that closely resembles the tolerized macrophage state, and this is compounded by a weak corticosteroid response capacity that may explain the lack of positive long-term effects of corticosteroid treatment for osteoarthritis patients.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Ruben De Francesco
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lieke A Lamers
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Sybren Rinzema
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Siebren Frölich
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Colin Logie
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Martijn H J van den Bosch
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
36
|
Islam K, Sancho-Shimizu V, Kampmann B, Diavatopoulos D, Holder B, Rice TF. Heterologous Effects of Pertussis and Influenza Vaccines During Pregnancy on Maternal and Infant Innate Immune Responses: A Pilot Study. Pediatr Infect Dis J 2025; 44:S70-S74. [PMID: 39951361 DOI: 10.1097/inf.0000000000004676] [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] [Indexed: 02/16/2025]
Abstract
BACKGROUND Research has demonstrated that some vaccines may have effects on the immune system beyond their intended targets. These heterologous effects of vaccination occur through reprogramming of innate immune cells, resulting in enhanced cytokine responses to unrelated pathogens and have been observed most evidently following Bacillus Calmette-Guérin vaccination. Pregnant women in the United Kingdom are offered influenza and acellular pertussis (Tdap) vaccines to protect the mother and infant, respectively, from infection. Little is known about the potential heterologous effects of vaccines given during pregnancy on the maternal and infant immune systems. OBJECTIVE To investigate heterologous innate immune responses in mothers and infants from pertussis-vaccinated and pertussis/influenza double-vaccinated pregnancies compared with unvaccinated pregnancies, in a pilot cohort. METHODS In this pilot study, samples collected as part of 2 maternal immunization studies were utilized. Maternal and cord peripheral blood mononuclear cells (PBMCs) were collected at birth from women who had received both Tdap and influenza vaccination, only the Tdap vaccine or no vaccines during pregnancy. To further investigate the effect of influenza vaccination alone, PBMCs were collected from nonpregnant women before and after seasonal influenza vaccination. PBMCs were incubated with pattern recognition receptor (PRR) ligands, vaccine adjuvants or CRM197 for 24 hours and cytokine responses were quantified in supernatants by enzyme-linked immunosorbent assay. RESULTS PBMC from women who received both Tdap and influenza vaccines had reduced IL-1β, IL-6 and IL-8 cytokine responses to PRR ligand stimulation, compared with those from women who received Tdap alone. Maternal vaccine status during pregnancy did not impact cytokine responses to PRR stimulation in cord PBMCs. Seasonal influenza vaccination did not alter cytokine responses to PRR ligands in nonpregnant women. CONCLUSIONS Our pilot study suggests that PBMC from women receiving combined Tdap and influenza vaccination during pregnancy may have reduced in vitro cytokine responses to nonpertussis stimuli. Larger cohorts of mother-infant pairs need to be studied to confirm these findings, study the potential mechanisms and control for potential confounders.
Collapse
Affiliation(s)
- Khaleda Islam
- From the Department of Metabolism, Digestion and Reproduction, Imperial College London
| | - Vanessa Sancho-Shimizu
- Department of Paediatric Infectious Diseases and Virology, Imperial College London, London, United Kingdom
| | - Beate Kampmann
- Centre for Global Health, Charité Universitatsmedizin, Berlin, Germany
| | - Dimitri Diavatopoulos
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Beth Holder
- From the Department of Metabolism, Digestion and Reproduction, Imperial College London
| | - Thomas F Rice
- Centre for Endocrinology, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
37
|
Bahl A, Pandey S, Rakshit R, Kant S, Tripathi D. Infection-induced trained immunity: a twist in paradigm of innate host defense and generation of immunological memory. Infect Immun 2025; 93:e0047224. [PMID: 39655962 PMCID: PMC11784091 DOI: 10.1128/iai.00472-24] [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: 02/01/2025] Open
Abstract
In contrast to adaptive immunity, which relies on memory T and B cells for long-term pathogen-specific responses, trained immunity involves the enhancement of innate immune responses through cellular reprogramming. Experimental evidence from animal models and human studies supports the concept of trained immunity and its potential therapeutic applications in the development of personalized medicine. However, there remains a huge gap in understanding the mechanisms, identifying specific microbial triggers responsible for the induction of trained immunity. This underscores the importance of investigating the potential role of trained immunity in redefining host defense and highlights future research directions. This minireview will provide a comprehensive summary of the new paradigm of trained immunity or innate memory pathways. It will shed light on infection-induced pathways through non-specific stimulation within macrophages and natural killer cells, which will be further elaborated in multiple disease perspectives caused by infectious agents such as bacteria, fungi, and viruses. The article further elaborates on the biochemical and cellular basis of trained immunity and its impact on disease status during recurrent exposures. The review concludes with a perspective segment discussing potential therapeutic benefits, limitations, and future challenges in this area of study. The review also sheds light upon potential risks involved in the induction of trained immunity.
Collapse
Affiliation(s)
- Aayush Bahl
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Saurabh Pandey
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, Delhi, India
| | - Roopshali Rakshit
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sashi Kant
- Bacterial Pathogenesis, Boehringer Ingelheim Animal Health USA Inc, Ames, Iowa, USA
| | - Deeksha Tripathi
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
38
|
Guo C, Xu P, Luo W, Zhang J, Sun X, Hoang H, Ma D, Wu D, Zhong J, Miao C. The Role of Dectin-1-Akt-RNF146 Pathway in β-Glucan Induced Immune Trained State of Monocyte in Sepsis. J Inflamm Res 2025; 18:1147-1165. [PMID: 39881796 PMCID: PMC11775823 DOI: 10.2147/jir.s482213] [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: 08/02/2024] [Accepted: 01/12/2025] [Indexed: 01/31/2025] Open
Abstract
Background Sepsis is regarded as a dysregulated immune response to infections. Recent study showed partially reversal of immunosuppression by trained immunity, which fosters an enhanced immune response towards a secondary challenge. However, the role of trained immunity in sepsis has not been fully understood. Methods We profiled the characteristics of peripheral blood mononuclear cells from septic patients using single-cell RNA sequencing (scRNA-seq) analyses. Murine double-hit models (pretreatment or post-treatment of β-glucan in septic mice) and murine monocyte/macrophage cell line RAW264.7 were used then. Results scRNA-seq revealed that Ring finger protein 146 (RNF146) and protein kinase B (Akt) were downregulated in the immunosuppression period of septic patients and were verified to be decreased in bone marrow and spleen monocytes from septic mice. While β-glucan pretreatment improved the immunosuppressed state in septic mice and increased dectin-1/Akt/RNF146 expressions in monocytes, along with the increased survival rate, inflammatory factors and aerobic glycolysis, indicating a change from immunosuppression to immune training. Moreover, RNF146 regulated dectin-1-Akt-mTOR signaling in the trained immune state of murine monocyte/macrophage RAW264.7 cell line and the expression of RNF146 was dependent on dectin-1-Akt activation. The inhibition of dectin-1 by its antagonist laminarin downregulated Akt-RNF146 signaling and partially reversed β-glucan induced trained immunity in septic mice. Conclusion RNF146 and Akt are downregulated in the immunosuppression period of sepsis, while increased after β-glucan pretreatment induced trained immunity in septic mice. Moreover, RNF146 regulates the immune trained state of monocyte through dectin-1-Akt-mTOR pathway, suggesting a possible target in reversal of immunosuppression in sepsis.
Collapse
Affiliation(s)
- Chenyue Guo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Peiyao Xu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Wenchen Luo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Jinlin Zhang
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Xingfeng Sun
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Harry Hoang
- Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Dehua Wu
- Department of Anesthesiology, Shanghai Songjiang District Central Hospital, Shanghai, People’s Republic of China
| | - Jing Zhong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| |
Collapse
|
39
|
Liao B, Zhang C, Shen J, Chen D, Wang J, Chen X, Zhou Y, Wei Y, Shi Y, Gou L, Guo Q, Zhou X, Xie H, Zhao L, Liao G, Zhu Z, Cheng L, Zhou X, Li Y, Ren B. Aloin remodels the cell wall of Candida albicans to reduce its hyphal virulence against oral candidiasis. Appl Microbiol Biotechnol 2025; 109:21. [PMID: 39853490 PMCID: PMC11761986 DOI: 10.1007/s00253-025-13411-7] [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: 11/15/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025]
Abstract
Aloe vera (L.) Burm.f. is a traditional Chinese medicine known for treating various ailments, including fungal infections. Aloin is one of the major components from A. vera, but its antifungal mechanism and therapeutic potential against oral candidiasis are not clear. This study aimed to examine the mechanism of aloin against Candida albicans and its inhibitory activity against oral candidiasis. In this study, we for the first time found that aloin could induce the formation of abnormal hyphae with smaller hyphal diameters and fewer branching points in C. albicans including 11 clinical isolates without growth inhibition. The transcriptome and further cell wall contents analysis indicated that aloin remodeled the cell wall to increase the contents of β-1,3-glucan and furtherly showed an antagonistic effect with micafungin. Aloin also significantly inhibited the cell damage of oral epithelial cells and oral candidiasis in mice infected by C. albicans due to its inhibitory actions on the hyphal development and expressions of virulence factors, including candidalysin (coded by ECE1). Our results suggest that aloin is a promising antifungal agent for controlling candidiasis and targeting hyphal development and pathogenesis represents a practical strategy for developing new antifungal drugs. KEY POINTS: • Aloin remodels the C. albicans cell wall to form avirulent hyphae. • Aloin inhibits C. albicans infections in oral epithelial cells and mouse mucosa without toxicity. • Aloin is a promising antifungal agent with therapeutic potential against C. albicans infections.
Collapse
Affiliation(s)
- Binyou Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chuanli Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiawei Shen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ding Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xi Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuan Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Wei
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lichen Gou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiang Guo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hongyu Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lin Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ga Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Information Management & Department of Stomatology Informatics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhuoli Zhu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Biao Ren
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
40
|
Mutchler AL, Haynes AP, Saleem M, Jamison S, Khan MM, Ertuglu L, Kirabo A. Epigenetic Regulation of Innate and Adaptive Immune Cells in Salt-Sensitive Hypertension. Circ Res 2025; 136:232-254. [PMID: 39819017 PMCID: PMC11750173 DOI: 10.1161/circresaha.124.325439] [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] [Indexed: 01/19/2025]
Abstract
Access to excess dietary sodium has heightened the risk of cardiovascular diseases, particularly affecting individuals with salt sensitivity of blood pressure. Our research indicates that innate antigen-presenting immune cells contribute to rapid blood pressure increases in response to excess sodium intake. Emerging evidence suggests that epigenetic reprogramming, with subsequent transcriptional and metabolic changes, of innate immune cells allows these cells to have a sustained response to repetitive stimuli. Epigenetic mechanisms also steer T-cell differentiation in response to innate immune signaling. Immune cells respond to environmental and nutritional cues, such as salt, promoting epigenetic regulation changes. This article aims to identify and discuss the role of epigenetic mechanisms in the immune system contributing to salt-sensitive hypertension.
Collapse
Affiliation(s)
- Ashley L. Mutchler
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexandria Porcia Haynes
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohammad Saleem
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mohd Mabood Khan
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lale Ertuglu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health
| |
Collapse
|
41
|
Das S, Lavine KJ. Role of Trained Immunity in Heath and Disease. Curr Cardiol Rep 2025; 27:18. [PMID: 39804563 DOI: 10.1007/s11886-024-02167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 05/02/2025]
Abstract
PURPOSE OF REVIEW This review aims to explore the role of immune memory and trained immunity, focusing on how innate immune cells like monocytes, macrophages, and natural killer cells undergo long-term epigenetic and metabolic rewiring. Specifically, it examines the mechanisms by which trained immunity, often triggered by infection or vaccination, could impact cardiac processes and contribute to both protective and pathological responses within the cardiovascular system. RECENT FINDINGS Recent research demonstrates that vaccination and infection not only activate immune responses in circulating monocytes and tissue macrophages but also affect immune progenitor cells within the bone marrow environment, conferring lasting protection against heterologous infections. These protective effects are attributed to epigenetic and metabolic reprogramming, which enable a heightened immune response upon subsequent encounters with pathogens. However, while trained immunity is beneficial in combating infections, it has been linked to exacerbated inflammation, which may increase susceptibility to cardiovascular diseases, including atherosclerosis and heart failure. Our review highlights the dual nature of trained immunity: while it offers protective advantages against infections, it also poses potential risks for cardiovascular health by promoting chronic inflammation. Understanding the molecular mechanisms underlying immune memory's impact on cardiac processes could lead to new therapeutic strategies to mitigate cardiovascular diseases, such as atherosclerosis, heart failure, and diabetes. These insights build the grounds for future research to balance the benefits of trained immunity with its potential risks in cardiovascular disease management.
Collapse
Affiliation(s)
- Shibali Das
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA.
| |
Collapse
|
42
|
Song X, Lei T, Cui N, Jin X, Huang Y, Shi Y, Zhao Z. A preliminary investigation on the protective effects of β-glucan and mannan induced trained immunity in pufferfish Takifugu obscurus. FISH & SHELLFISH IMMUNOLOGY 2025; 156:110035. [PMID: 39577788 DOI: 10.1016/j.fsi.2024.110035] [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: 08/25/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Immune stimuli are able to trigger long-term protective effects through mechanisms of trained immunity, which has attracted increasing attention. Although the existence of trained immunity has evidenced in teleost fish, while there were no such reports in pufferfish (Takifugu obscurus) so far. Therefore, the present study aimed to evaluate the induction of β-glucan and mannan on the trained immunity and their protective efficacy against Vibrio harveyi re-stimulation in pufferfish. β-glucan and mannan induction of trained immunity in head-kidney primary leukocytes is accompanied by a strong increase in immediate ROS burst, cumulative NO production and lactate concentrations after V. harveyi re-stimulation. In addition, β-glucan and mannan-treated pufferfish exhibited reduced bacterial loads in multiple tissues, a rapid and long-term elevated inflammatory response in head kidney during secondary V. harveyi infection. Notably, immune receptors dectin-1 and dectin-2, and cytokines tnfsf14 and il-1β exhibited comparatively upregulation to the β-glucan training, while NK-lysin and faslg showed stronger response to the mannan training post V. harveyi stimulation, implying the different signaling pathway activated post β-glucan and mannan training. Subsequent markers for immune training including abundance of genes encoding glycolytic enzymes (hk1, pfkla, and ldha) and transcription factors (mtor and hif-1α), as well as increased acetylation levels were elevated in the β-glucan and mannan trained pufferfish, depicting heightened glycolysis following β-glucan and mannan training. These results collectively demonstrated that β-glucan and mannan both induced protective responses against V. harveyi infection probably through mediating distinct signaling pathway in pufferfish, and studies are underway to harness its potential applicability for prime and boost vaccination strategies.
Collapse
Affiliation(s)
- Xiaorui Song
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Tianying Lei
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Nan Cui
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Xingkun Jin
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Ying Huang
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Yan Shi
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Zhe Zhao
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China.
| |
Collapse
|
43
|
Jiang W, Chen Y, Yu CY, Zou B, Lu Y, Yang Q, Tang Z, Mao W, Li J, Han H, Shao L, Zeng J, Chu Y, Tang J, Lu M. Alveolar epithelial cells shape lipopolysaccharide-induced inflammatory responses and reprogramming of alveolar macrophages. Eur J Immunol 2025; 55:e2350378. [PMID: 39498697 DOI: 10.1002/eji.202350378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 11/07/2024]
Abstract
Alveolar macrophages (AMs) are sentinels in the airways, where they sense and respond to invading microbes and other stimuli. Unlike macrophages in other locations, AMs can remain responsive to Gram-negative lipopolysaccharides (LPS) after they have responded to LPS in vivo (they do not develop "endotoxin tolerance"), suggesting that the alveolar microenvironment may influence their responses. Although alveolar epithelial cells (AECs) normally limit AMs' innate responses, preventing inflammation induced by harmless antigens in the lung, how AECs influence the innate responses of AMs to infectious agents has been uncertain. Here we report that (1) after exposure to aspirated (intranasal instillation) LPS, AMs increase their responses to TLR agonists and elevate their phagocytic and bactericidal activities in mice; (2) Aspirated LPS pre-exposure increases host resistance to pulmonary infection caused by Gram-negative bacteria and the protection effect lasts for at least 35 days; (3) LPS stimulation of AECs both increases AMs' innate immune responses and prevents AMs from developing tolerance in vitro; (4) Upon LPS stimulation, AMs secreted TNF-α induces AECs to release GM-CSF, which potentiates AMs' response. These experiments have revealed a previously unappreciated role that AECs may play in boosting the innate responses of AMs and promoting resistance to pulmonary infections.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Department of Rheumatology and Immunology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yeying Chen
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Cheng-Yun Yu
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Benkun Zou
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yimeng Lu
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Qian Yang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Zihui Tang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiying Mao
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Li
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Han Han
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Lingyun Shao
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiashun Zeng
- Department of Rheumatology and Immunology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Mingfang Lu
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
| |
Collapse
|
44
|
Mao H, Liu Y, Lv Q, Li C, Yang Y, Wu F, Xu N, Jin X. The effect of β-Glucan induced intestinal trained immunity against Trichinella spiralis infection. Vet Parasitol 2025; 333:110238. [PMID: 38944590 DOI: 10.1016/j.vetpar.2024.110238] [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: 04/22/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/01/2024]
Abstract
Parasitic helminth Trichinella spiralis (Ts) induce mixed Th1/Th2 response with predominant type 2 immune responses, with protective immunity mediated by interleukin (IL)-4, IL-5, and IL-13. β-Glucan (BG) has been shown to have the ability to induce trained immunity, confers non-specific protection from secondary infections. However, whether BG-induced trained immunity played a role in protective type 2 immunity against Ts infection is unclear. In this study, BG was administered five days before Ts infection to induce trained immunity. Our findings demonstrate that BG pretreatment effectively reduced the number of T. spiralis adults and muscle larvae, whereas inhibition of trained immunity abolished the effect of BG. Additionally, we observed a significant increase in goblet cells and mucus production as evidenced by Alcian blue periodic acid-Schiff staining. Furthermore, quantitative real-time PCR analysis revealed a significant upregulation of IL-4, IL-5, and IL-13 expression in response to BG. Conversely, the inhibitor of trained immunity reversed these effects, suggesting that BG-induced trained immunity confers strong protection against Ts infection. In conclusion, these findings suggest that BG-induced trained immunity may play a role in protection against infections caused by other helminths.
Collapse
Affiliation(s)
- Hanhai Mao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yi Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qingbo Lv
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chengyao Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yaming Yang
- Department of Helminth, Yunnan Institute of Parasitic Diseases, Puer, China
| | - Fangwei Wu
- Department of Helminth, Yunnan Institute of Parasitic Diseases, Puer, China
| | - Ning Xu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
45
|
Tehrani SSH, Kogan A, Mikulski P, Jansen LET. Remembering foods and foes: emerging principles of transcriptional memory. Cell Death Differ 2025; 32:16-26. [PMID: 37563261 PMCID: PMC11748651 DOI: 10.1038/s41418-023-01200-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/20/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
Transcriptional memory is characterized by a primed cellular state, induced by an external stimulus that results in an altered expression of primed genes upon re-exposure to the inducing signal. Intriguingly, the primed state is heritably maintained across somatic cell divisions even after the initial stimulus and target gene transcription cease. This phenomenon is widely observed across various organisms and appears to enable cells to retain a memory of external signals, thereby adapting to environmental changes. Signals range from nutrient supplies (food) to a variety of stress signals, including exposure to pathogens (foes), leading to long-term memory such as in the case of trained immunity in plants and mammals. Here, we review these priming phenomena and our current understanding of transcriptional memory. We consider different mechanistic models for how memory can work and discuss existing evidence for potential carriers of memory. Key molecular signatures include: the poising of RNA polymerase II machinery, maintenance of histone marks, as well as alterations in nuclear positioning and long-range chromatin interactions. Finally, we discuss the potential adaptive roles of transcriptional memory in the organismal response to its environment from nutrient sensing to trained immunity.
Collapse
Affiliation(s)
- Sahar S H Tehrani
- Department of Biochemistry, University of Oxford, OX1 3QU, Oxford, UK
| | - Anna Kogan
- Department of Biochemistry, University of Oxford, OX1 3QU, Oxford, UK
| | - Pawel Mikulski
- Department of Biochemistry, University of Oxford, OX1 3QU, Oxford, UK.
| | - Lars E T Jansen
- Department of Biochemistry, University of Oxford, OX1 3QU, Oxford, UK.
| |
Collapse
|
46
|
Angulo M, Angulo C. Trained immunity-based vaccines: A vision from the one health initiative. Vaccine 2025; 43:126505. [PMID: 39520776 DOI: 10.1016/j.vaccine.2024.126505] [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: 08/16/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Trained immunity-based vaccines (TIbV or TRIMbV) represent a novel approach to combating infectious diseases. The innate immune system in animals, including humans, exhibits "memory-like" functions. Remarkably, the immunological mechanisms -both epigenetic and metabolic-) underlying this memory enables immune cells to develop defensive and protective outcomes against unspecific pathogenic infections. Under this context, the One Health initiative promotes integrative efforts to combat zoonotic (and anthropozoonotic) diseases, which is critical because 3 of 4 animal infections are transmitted to humans. Therefore, TIbV constitutes a potential affordable approach to control zoonotic pathologies, especially under pandemic scenarios. This review describes the state-of-the-art TIbV and their hurdles, opportunities, and prospects for the One Health initiative to prevent, control, and treat infectious diseases.
Collapse
Affiliation(s)
- Miriam Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD), Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S., C.P. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD), Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S., C.P. 23096, Mexico.
| |
Collapse
|
47
|
Liu J, Tian H, Ju J, Nie F, Yin Q, Zhao J, Wang S, Guo H, Yang P. Porphyromonas gingivalis-Lipopolysaccharide Induced Gingival Fibroblasts Trained Immunity Sustains Inflammation in Periodontitis. J Periodontal Res 2024. [PMID: 39665166 DOI: 10.1111/jre.13372] [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: 09/20/2024] [Revised: 11/07/2024] [Accepted: 11/17/2024] [Indexed: 12/13/2024]
Abstract
AIM To investigate whether trained immunity occurs in gingival fibroblasts (GFs) and its relationship to the persistence of inflammation in periodontitis. METHODS Periodontally healthy and inflammatory gingival fibroblasts (HGFs and IGFs) were cultured through continuous adherence subculture of tissue blocks. Trained immunity in HGFs was evaluated via a classic in vitro model, with relevant markers assessed via enzyme-linked immunosorbent assay, lactate content assay, glycolytic rate assay, and chromatin immunoprecipitation. A histone methyltransferase blocker and a PI3K inhibitor were added to investigate the mechanisms underlying trained immunity. The relationship between trained immunity and periodontitis was further examined via immunofluorescence staining and chromatin immunoprecipitation on IGFs. RESULTS Compared with untrained cells, GFs trained with Porphyromonas gingivalis-lipopolysaccharide (P. gingivalis-LPS) exhibited a significant increase in IL-6 and TNF-α secretion, enhanced glycolytic metabolism, and enriched mono-methylation of lysine 4 on histone H3 (H3K4me1) at the enhancer regions of TNF-α and IL-6. The addition of a histone methyltransferase blocker and a PI3K inhibitor greatly reduced trained immunity. Additionally, the response of IGFs to P. gingivalis-LPS stimulation and their epigenetic modifications were similar to those observed in trained HGFs. CONCLUSION This study novelly discovered that both P. gingivalis-LPS-stimulated HGFs and IGFs in periodontitis acquired trained immunity. Following P. gingivalis-LPS stimulation, HGFs underwent metabolic and epigenetic changes via the PI3K/AKT pathway, with these epigenetic changes also observed in IGFs. This finding suggests that trained immunity in GFs may be a key mechanism underlying the recurrence and persistence of periodontitis.
Collapse
Affiliation(s)
- Jiayi Liu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Haoyang Tian
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Jinhong Ju
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Fujiao Nie
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Qiuyue Yin
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Jingjing Zhao
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Suli Wang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Hongmei Guo
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Pishan Yang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| |
Collapse
|
48
|
Ruffinatto L, Groult Y, Iacono J, Sarrazin S, de Laval B. Hematopoietic stem cell a reservoir of innate immune memory. Front Immunol 2024; 15:1491729. [PMID: 39720722 PMCID: PMC11666435 DOI: 10.3389/fimmu.2024.1491729] [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: 09/05/2024] [Accepted: 10/30/2024] [Indexed: 12/26/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are a rare, long-lived and multipotent population that give rise to majority of blood cells and some tissue-resident immune cells. There is growing evidence that inflammatory stimuli can trigger persistent reprogramming in HSCs that enhances or inhibits the cellular functions of these HSCs and their progeny in response to subsequent infections. This newly discovered property makes HSCs a reservoir for innate immune memory. The molecular mechanisms underlying innate immune memory in HSCs are similar to those observed in innate immune cells, although their full elucidation is still pending. In this review, we examine the current state of knowledge on how an inflammatory response leads to reprogramming of HSCs. Understanding the full spectrum of consequences of reshaping early hematopoiesis is critical for assessing the potential benefits and risks under physiological and pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Bérengère de Laval
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut
National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
49
|
Tamburini B, Sheridan R, Doan T, Lucas C, Forward T, Fleming I, Uecker-Martin A, Morrison T, Hesselberth J. A specific gene expression program underlies antigen archiving by lymphatic endothelial cells in mammalian lymph nodes. RESEARCH SQUARE 2024:rs.3.rs-5493746. [PMID: 39711554 PMCID: PMC11661310 DOI: 10.21203/rs.3.rs-5493746/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Lymph node (LN) lymphatic endothelial cells (LEC) actively acquire and archive foreign antigens. Here, we address questions of how LECs achieve durable antigen archiving and whether LECs with high levels of antigen express unique transcriptional programs. We used single cell sequencing in dissociated LN tissue and spatial transcriptomics to quantify antigen levels in LEC subsets and dendritic cell populations at multiple time points after immunization and determined that ceiling and floor LECs archive antigen for the longest duration. We identify, using spatial transcriptomics, antigen positive LEC-dendritic cell interactions. Using a prime-boost strategy we find increased antigen levels within LECs after a second immunization demonstrating that LEC antigen acquisition and archiving capacity can be improved over multiple exposures. Using machine learning we defined a unique transcriptional program within archiving LECs that predicted LEC archiving capacity in mouse and human independent data sets. We validated this modeling, showing we could predict lower levels of LEC antigen archiving in chikungunya virus-infected mice and demonstrated in vivo the accuracy of our prediction. Collectively, our findings establish unique properties of LECs and a defining transcriptional program for antigen archiving that can predict antigen archiving capacity in different disease states and organisms.
Collapse
Affiliation(s)
| | | | - Thu Doan
- University of Colorado Anschutz Medical Campus
| | | | | | | | | | | | | |
Collapse
|
50
|
Rai MN, Rai R. H 3K 4 Methylation and Demethylation in Fungal Pathogens: The Epigenetic Toolbox for Survival and Adaptation in the Host. Pathogens 2024; 13:1080. [PMID: 39770340 PMCID: PMC11728789 DOI: 10.3390/pathogens13121080] [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: 09/25/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 01/14/2025] Open
Abstract
Pathogenic fungi represent a diverse group of eukaryotic microorganisms that significantly impact human health and agriculture. In recent years, the role of epigenetic modifications, particularly histone modifications, in fungal pathobiology has emerged as a prominent area of interest. Among these modifications, methylation of histone H3 at lysine-4 (H3K4) has garnered considerable attention for its implications in regulating gene expression associated with diverse cellular processes. A body of literature has uncovered the pivotal roles of H3K4 methylation in multiple biological processes crucial for pathogenic adaptation in a wide range of fungal pathogens of humans and food crops. This review delves into the recent advancements in understanding the impact of H3K4 methylation/demethylation on fungal pathogenesis. We explore the roles of H3K4 methylation in various cellular processes, including fungal morphogenesis and development, genome stability and DNA repair, metabolic adaptation, cell wall maintenance, biofilm formation, antifungal drug resistance, and virulence. We also discuss the conservation of H3K4 methylation regulators and their potential as therapeutic targets to prevent fungal diseases. Collectively, this review underscores the intricate links between H3K4 methylation, fungal pathogenesis, and potential avenues for novel antifungal strategies.
Collapse
Affiliation(s)
- Maruti Nandan Rai
- College of Agricultural, Consumer, and Environmental Sciences (ACES), University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Rikky Rai
- Department of Botany, University of Allahabad, Prayagraj 211002, Uttar Pradesh, India;
| |
Collapse
|