1
|
Lane JI, Nieves-Ortiz E, Ndatabaye O, Fatkhullina AR, Lopez S, Dermody TS, Esterházy D. Intestinal lymphatic vasculature is functionally adapted to different drainage regions and is altered by helminth infection. J Exp Med 2025; 222:e20241181. [PMID: 40505102 PMCID: PMC12162095 DOI: 10.1084/jem.20241181] [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: 07/09/2024] [Revised: 03/18/2025] [Accepted: 05/12/2025] [Indexed: 06/16/2025] Open
Abstract
We sought to determine whether the lymphatic vasculature functionally adapts to the organ in which it resides, such as along the gut. Duodenal lymphatic capillaries (lacteals) displayed the most discontinuous tight junction composition within the gut, resulting in a dependence on duodenal lacteals for rapid dietary lipid uptake. Duodenal helminths abrogated these features. Parallel RNA sequencing of lymphatic endothelial cells and mucosa along the intestine revealed that the transcriptomes overlapped in functional profiles. RNA sequencing also identified a putative VEGFR-2/3 signaling gradient that may explain differences in lacteal tight junctions along the small intestine at homeostasis. Transcriptionally, helminth infection triggered antimicrobial and angiogenic responses. While microbial depletion acted additively to helminths on lymphatic restructuring, glucocorticoids partially reversed helminth-induced lacteal changes. This suggests helminths induce lymphangiogenesis and associated lymphatic "zippering" via inflammation. Our study uncovers and explains the superior lipid absorption by duodenal lacteals and how it is compromised by helminths and provides transcriptional insights into lymphatic function along the gut.
Collapse
Affiliation(s)
- Jorden I. Lane
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | | | | | | - Sebastian Lopez
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Terence S. Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Daria Esterházy
- Department of Pathology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Kreslavsky T. Thymflammation: The Role of a Constitutively Active Inflammatory Network and "Ectopic" Cell Types in the Thymus in the Induction of T Cell Tolerance and Beyond. Immunol Rev 2025; 332:e70037. [PMID: 40433806 PMCID: PMC12117520 DOI: 10.1111/imr.70037] [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: 01/31/2025] [Revised: 03/28/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
The thymus exhibits constitutive activation of nearly all major inflammatory pathways, including sterile MyD88-dependent signaling and interferon production by mTECs, the presence of cellular and molecular components of type 1, type 2, and type 3 responses, as well as sustained B cell activation. The reasons for the existence of such a complex constitutively active inflammatory network at the site of T cell development-where the initial pathogen encounter is unlikely-have remained enigmatic. We propose that this inflammatory thymic 'ecosystem' has evolved to promote immunological tolerance to 'inflammatory self'-endogenous molecules absent from most peripheral tissues at steady state but upregulated during pathogen invasion. The spatial and temporal overlap with pathogen presence makes the discrimination of the inflammatory self from pathogen-derived molecules a unique challenge for the adaptive immune system. The frequent occurrence of diseases associated with autoantibodies against proinflammatory cytokines underscores the persistent risk of these molecules being misidentified as foreign. Their abundant representation in the thymus, therefore, is likely to be critical for maintaining tolerance. This review explores current insights into the thymic inflammatory network, its cellular and molecular constituents, and their role in the induction of T cell tolerance.
Collapse
Affiliation(s)
- Taras Kreslavsky
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| |
Collapse
|
3
|
Chi Y, Jiang H, Yin Y, Zhou X, Shao Y, Li Y, Rao J. Macrophage Signaling Pathways in Health and Disease: From Bench to Bedside Applications. MedComm (Beijing) 2025; 6:e70256. [PMID: 40529613 PMCID: PMC12171086 DOI: 10.1002/mco2.70256] [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: 09/24/2024] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 06/20/2025] Open
Abstract
Macrophages exhibit remarkable functional plasticity by dynamically polarizing into proinflammatory or antiinflammatory subsets in response to microenvironmental cues. This duality underpins their pivotal roles in immune defense, tissue homeostasis, and disease progression; however, the molecular mechanisms governing their polarization and crosstalk across various pathologies remain incompletely defined. This review systematically delineates macrophage biology, emphasizing the interplay between subset-specific signaling networks and their context-dependent activation in both health and disease. The heterogeneity of macrophages is characterized by detailing the distinctions between tissue-resident and monocyte-derived origins, as well as their polarization states. Core pathways regulating phagocytosis, tissue repair, immune modulation, and neuroprotection are dissected, along with their dysregulation in autoimmune disorders, neurodegeneration, cancers, and cardiovascular diseases. Notably, microenvironmental factors such as damage-associated molecular patterns, pathogen-associated molecular patterns, and metabolic intermediates dynamically reshape macrophage phenotypes through NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome activation or signal transducer and activator of transcription (STAT)-mediated transcriptional control. Preclinical and clinical evidence underscores potential therapeutic targets and emerging strategies. The significance of this review lies in its integrative analysis of signaling crosstalk, paradoxical pathway roles, and translational implications for precision therapies. These insights into macrophage functions and signaling pathways provide a robust foundation for future disease intervention and personalized medicine.
Collapse
Affiliation(s)
- Yongquan Chi
- Hepatobiliary CenterKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Hepatobiliary CancersThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Haipeng Jiang
- Hepatobiliary CenterKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Hepatobiliary CancersThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Yiyuan Yin
- Nanjing Medical UniversityNanjingJiangsuChina
| | - Xinyu Zhou
- Nanjing Medical UniversityNanjingJiangsuChina
| | | | - Yongsheng Li
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| | - Jianhua Rao
- Hepatobiliary CenterKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Hepatobiliary CancersThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
4
|
Erjefält JS. Spatial Eosinophil Phenotypes as Immunopathogenic Determinants in Inflammatory Diseases. Cells 2025; 14:847. [PMID: 40498023 PMCID: PMC12154561 DOI: 10.3390/cells14110847] [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: 03/23/2025] [Revised: 05/27/2025] [Accepted: 05/28/2025] [Indexed: 06/19/2025] Open
Abstract
Eosinophils are increasingly recognized as adaptable immune cells that exhibit diverse phenotypes and effector functions across different tissues and disease states. While they can induce pathology through degranulation and cytotoxic mediator release, eosinophils also fulfill regulatory and tissue repair roles. Advances in single-cell and spatial technologies have begun to reveal how microenvironmental cues (including cytokines, chemokines, and cell-cell interactions) shape eosinophil behavior in health and disease. These insights are critical for understanding why certain patients respond variably to therapies targeting eosinophils and related type 2 pathways. By dissecting eosinophil heterogeneity in real human tissues, researchers may identify new biomarkers, refine endotyping approaches, and develop more precise therapeutic strategies. This review summarizes emerging concepts of eosinophil biology in inflammatory conditions, highlights the impact of spatial context on eosinophil functions, and discusses the future of advanced phenotyping in guiding personalized treatments.
Collapse
Affiliation(s)
- Jonas S. Erjefält
- Unit of Airway Inflammation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden;
- Department of Respiratory Medicine and Allergology, Lund University, 221 85 Lund, Sweden
| |
Collapse
|
5
|
Kibet M, Abebayehu D. Crosstalk between T cells and fibroblasts in biomaterial-mediated fibrosis. Matrix Biol Plus 2025; 26:100172. [PMID: 40226302 PMCID: PMC11986236 DOI: 10.1016/j.mbplus.2025.100172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Biomaterial implants are a critical aspect of our medical therapies and biomedical research and come in various forms: stents, implantable glucose sensors, orthopedic implants, silicone implants, drug delivery systems, and tissue engineered scaffolds. Their implantation triggers a series of biological responses that often times lead to the foreign body response and subsequent fibrotic encapsulation, a dense ECM-rich capsule that isolates the biomaterial and renders it ineffective. These responses lead to the failure of biomaterials and is a major hurdle to overcome and in promoting their success. Much attention has been given to macrophage populations for the inflammatory component of these responses to biomaterials but recent work has identified an important role of T cells and their ability to modulate fibroblast activity and vice versa. In this review, we focus on T cell-fibroblast crosstalk by exploring T cell subsets, critical signaling pathways, and fibroblast populations that have been shown to dictate biomaterial-mediated fibrosis. We then highlight emerging technologies and model systems that enable new insights and avenues to T cell-fibroblast crosstalk that will improve biomaterial outcomes.
Collapse
Affiliation(s)
- Mathew Kibet
- Department of Biomedical Engineering, School of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, United States
| | - Daniel Abebayehu
- Department of Biomedical Engineering, School of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, United States
| |
Collapse
|
6
|
Horisaka H, Yokawa S, Suzuki R, Emoto R, Maeda R, Furuno T. Suppression of FcεRI-evoked Degranulation in RBL-2H3 Cells on Gelatin Methacryloyl Hydrogel. Cell Biochem Biophys 2025; 83:2481-2488. [PMID: 39731647 DOI: 10.1007/s12013-024-01657-3] [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: 12/18/2024] [Indexed: 12/30/2024]
Abstract
Cell-extracellular matrix (ECM) interactions play multiple roles in developmental, physiological, and pathological processes. ECM stiffness substantially affects cellular morphology, migration, and function. In this study, we investigated the effect of ECM comprising gelatin methacryloyl (GelMA) on the activation of rat basophilic leukemia (RBL-2H3) cells, a model mast cell line. Maintenance of intracellular Ca2+ concentration ([Ca2+]i) elevation and subsequent degranulation, evoked by crosslinking the high-affinity IgE receptors (FcεRI), were significantly suppressed in RBL-2H3 cells on collagen-coated GelMA hydrogel than those on collagen-coated glass dishes and plastic wells. Thapsigargin and phorbol myristate acetate caused sustained [Ca2+]i increase and degranulation to a similar extent in cells on both GelMA hydrogel and plastic wells/glass dishes. F-actin was clearly accumulated along the periphery of RBL-2H3 cells in plane attached to glass, but not GelMA hydrogel, suggesting that the loose actin cytoskeleton of RBL-2H3 cells on GelMA hydrogel caused suppressive degranulation through unstable FcεRI aggregation.
Collapse
Affiliation(s)
- Haruna Horisaka
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Satoru Yokawa
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Ruriko Suzuki
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Rin Emoto
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Rino Maeda
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Tadahide Furuno
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| |
Collapse
|
7
|
Tran LA, Catlin M, Schecter S, Thurman AL, Ghimire S, Tudas R, Bettis B, Gannon R, Zabner J, Pezzulo AA. The extracellular matrix protein periostin is required for wound repair in primary human airway epithelia. Am J Physiol Lung Cell Mol Physiol 2025; 328:L826-L831. [PMID: 40257107 PMCID: PMC12100768 DOI: 10.1152/ajplung.00039.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/24/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025] Open
Abstract
Type 2 inflammation and epithelial-to-mesenchymal transitions (EMTs) play critical roles in airway repair after damage from allergens or parasites. The matricellular protein periostin (POSTN) has increased expression in inflammatory conditions and has been implicated in fibrosis and EMT, suggesting a role in airway repair. This study investigates the role of periostin in airway epithelial and lung fibroblast wound repair using an in vitro wound model. Our results demonstrate that the type 2 cytokine IL-13 induces periostin secretion from primary human airway epithelial basal cells. Periostin knockdown in human airway epithelial cells (HAEs) and human lung fibroblasts (HLFs) impairs wound closure, indicating that periostin is required for airway repair. In a coculture model of HAE and HLFs, fibroblast-secreted POSTN is required for airway epithelial wound repair, suggesting that periostin is involved in paracrine signaling between the two cell types. These findings highlight periostin's critical function in epithelial and fibroblast-mediated wound repair, suggesting its potential as a therapeutic target for diseases characterized by aberrant wound healing and fibrosis, such as asthma and idiopathic pulmonary fibrosis.NEW & NOTEWORTHY This article highlights the critical role of periostin (POSTN) in airway epithelial and fibroblast-mediated wound repair. Moreover, the study reveals a paracrine signaling loop between airway epithelial basal cells and lung fibroblasts, emphasizing periostin's therapeutic potential for diseases like asthma and idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Lorena A. Tran
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Michael Catlin
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Scott Schecter
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Andrew L. Thurman
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Shreya Ghimire
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Rosarie Tudas
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Brandon Bettis
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Ryan Gannon
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Joseph Zabner
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Alejandro A. Pezzulo
- Department of Internal Medicine, Roy J. and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
8
|
Valenti G, Laise P, Wu F, Takahashi R, Ruan T, Vasciaveo A, Jiang Z, Kobayashi H, Sunagawa M, Middelhoff M, Nienhüser H, Fu N, Malagola E, Companioni O, Hayakawa Y, Iuga AC, Califano A, Wang TC. Regulatory network analysis of Dclk1 gene expression reveals a tuft cell-ILC2 axis that inhibits pancreatic tumor progression. Cell Rep 2025; 44:115734. [PMID: 40408246 DOI: 10.1016/j.celrep.2025.115734] [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: 11/14/2024] [Revised: 01/21/2025] [Accepted: 05/02/2025] [Indexed: 05/25/2025] Open
Abstract
Doublecortin-like kinase 1 (Dclk1) expression identifies cells that are rare in normal pancreas but occur with an increased frequency in pancreatic neoplasia. The identity of these cells has been a matter of debate. We employed Dclk1 reporter mouse models and single-cell RNA sequencing (scRNA-seq) to define Dclk1-expressing cells. In normal pancreas, Dclk1 identifies subsets of ductal, islet, and acinar cells. In pancreatic neoplasia, Dclk1 identifies several cell populations, among which acinar-to-ductal metaplasia (ADM)-like cells and tuft-like cells are predominant. These two populations play opposing roles, with Dclk1+ ADM-like cells sustaining and Dclk1+ tuft-like cells restraining tumor progression. The generation of Dclk1+ tuft-like cells requires the transcription factor SPIB and is sustained by a paracrine loop involving type 2 innate lymphoid cells (ILC2s) and cancer-associated fibroblasts (CAFs) that provide interleukin (IL)-13 and IL-33, respectively. Dclk1+ tuft-like cells release angiotensinogen to restrain tumor progression. Overall, our study defines pancreatic Dclk1+ cells and unveils a protective tuft cell-ILC2 axis against pancreatic neoplasia.
Collapse
Affiliation(s)
- Giovanni Valenti
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Pasquale Laise
- Department of Systems Biology, Columbia University, New York, NY, USA; DarwinHealth, Inc., New York, NY, USA
| | - Feijing Wu
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Ryota Takahashi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Tuo Ruan
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | | | - Zhengyu Jiang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Hiroki Kobayashi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Masaki Sunagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Moritz Middelhoff
- Klinik und Poliklinik für Innere Medizin II, Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Henrik Nienhüser
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Na Fu
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Osmel Companioni
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Yoku Hayakawa
- Graduate School of Medicine, Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Alina C Iuga
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA; DarwinHealth, Inc., New York, NY, USA; Chan Zuckerberg Biohub New York, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA; Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
9
|
Gholizadeh F, Hajiaghayi M, Choi JS, Little SR, Rahbari N, Kargar M, Brotto K, Han E, Shih SCC, Darlington PJ. Modulatory effects of M3 muscarinic acetylcholine receptor on inflammatory profiles of human memory T helper cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf086. [PMID: 40405417 DOI: 10.1093/jimmun/vkaf086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 03/24/2025] [Indexed: 05/24/2025]
Abstract
Memory T helper (Th) cells, generated in response to immunogenic challenges, are crucial in orchestrating adaptive immune responses. Acetylcholine (ACh), a key neurotransmitter of the parasympathetic nervous system, modulates immune function via muscarinic ACh receptors (mAChRs). This study investigates the role of mAChRs, particularly the M3 muscarinic ACh receptor (M3R), in regulating the cytokine and chemokine profile and NF-κB p65 activity in primary human memory Th cells. Memory Th cells were isolated from healthy donors and stimulated with anti-CD3/CD28/CD2 in the presence of oxotremorine-M (M1R-M5R agonist), atropine (M1R-M5R antagonist), or J104129 (M3R-selective antagonist). CHRM1-CHRM5 expression was quantified using RT-qPCR. M3R and phosphorylated NF-κB p65 were analyzed by Western blot. IFN-γ, IL-17A, and IL-4 were assessed by ELISA, while intracellular cytokine and chemokine receptor expression were measured by flow cytometry. CHRM3 knockout was performed using CRISPR-Cas9. Memory Th cells expressed all 5 mAChR subtypes. Oxotremorine-M increased IFN-γ and IL-17A while reducing IL-4 in an atropine-sensitive manner. Blocking or knocking out M3R prevented oxotremorine-M-induced increases in IFN-γ and IL-17A, but the suppression of IL-4 remained unchanged. Stimulation of mAChRs, particularly M3R, enhanced NF-κB p65 activity but did not affect chemokine receptor expression, cell proliferation, viability, or M3R levels. These findings indicate that mAChRs, including M3R, drive a pro-inflammatory memory Th-cell response through NF-κB p65 activation, while IL-4 suppression occurs independently of M3R. Targeting M3R specifically may provide a strategy for modulating adaptive immunity and treating inflammatory diseases.
Collapse
Affiliation(s)
| | - Mehri Hajiaghayi
- Department of Biology, Concordia University, Montréal, Québec, Canada
| | - Jennifer S Choi
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Samuel R Little
- Department of Electrical and Computer Engineering, Concordia University, Montréal, Québec, Canada
| | - Niloufar Rahbari
- Department of Chemical and Materials Engineering, Concordia University, Montréal, Québec, Canada
| | - Melika Kargar
- Department of Biology, Concordia University, Montréal, Québec, Canada
| | - Kelly Brotto
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Eric Han
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Steve C C Shih
- Department of Electrical and Computer Engineering, Concordia University, Montréal, Québec, Canada
| | - Peter J Darlington
- Department of Biology, Concordia University, Montréal, Québec, Canada
- Department of Health, Kinesiology & Applied Physiology, Concordia University, Montréal, Québec, 7141 Sherbrooke Street west, H4B 1R6, Canada
| |
Collapse
|
10
|
Salihah SS, Bibi B, Khan S, Tahir M, Mahmood S, Vosough M, Razzoli A, Sidoli S, Gul A. 3D Biomimetic Liver Cancer Model: Diethylnitrosamine-Induced Proteomic Dysregulations in Stromal-Epithelial Milieu. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.17.654447. [PMID: 40475465 PMCID: PMC12139896 DOI: 10.1101/2025.05.17.654447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2025]
Abstract
Hydrogel-based three-dimensional (3D) co-culture systems are emerging as biomimetic platforms that more accurately recapitulate tissue architecture and microenvironmental interactions compared to conventional two-dimensional (2D) cultures. This study introduces an engineered 3D liver-like model to investigate compartment-specific responses to the potent hepatocarcinogen Diethylnitrosamine (DEN), with a focus on early events in carcinogenesis and tumor-stroma interactions. AML12 and 3T3 cell lines were treated with DEN or vehicle either in 2D culture or in 3D hydrogels in four experimental groups: (1) DEN-treated AML12 with vehicle-treated 3T3, (2) DEN-treated 3T3 with vehicle-treated AML12, (3) both cell types DEN-treated, and (4) both vehicle-treated. The cultured recombinants were subjected to proteomic profiling via mass spectrometry, followed by bioinformatics analysis and the results were validated through immunocytochemical staining (ICC). Gene ontology analysis revealed that cytoskeletal, RNA metabolism, and scaffold/adaptor proteins were among the most significantly enriched in 3D versus 2D models. Structural proteins emerged exclusively in mixed 3D co-cultures, reinforcing the organotypic nature of the system. Enriched pathways in 3D included intermediate filament organization, actin dynamics, and focal adhesion-pathways closely associated with liver carcinogenesis. Protein-protein interaction analysis demonstrated maximal network complexity in 3D cultures where both compartments were DEN-exposed. Survival analysis further identified poor-prognosis biomarkers (KRT20, KRT15, KRT14) uniquely enriched in this condition. ICC staining supported the proteomic findings. This organoid-like 3D co-culture model provides a physiologically relevant platform for investigating early-stage liver carcinogenesis and highlights the critical role of stromal-epithelial interactions. Its ability to replicate organ-level complexity and generate clinically relevant proteomic signatures supports its utility in translational cancer research and future drug discovery applications.
Collapse
Affiliation(s)
- Salmma S Salihah
- Department of Biological Sciences, International Islamic University Islamabad, Pakistan
| | - Bareerah Bibi
- School of Interdisciplinary Engineering & Science (SINES), NUST, Islamabad, Pakistan
| | - Sehrish Khan
- Department of Biological Sciences, International Islamic University Islamabad, Pakistan
| | - Muhammad Tahir
- Biomedical Mass Spectrometry and Systems Biology, University of South Denmark, Odense, Denmark
- Present address: `Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Sana Mahmood
- Department of Biological Sciences, International Islamic University Islamabad, Pakistan
| | - Massoud Vosough
- Royan Institute for Stem Cell Biology & Regenerative Medicine
- Karolinska Institute | Experimental Cancer Medicine, Sweden
| | - Agnese Razzoli
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Itlay
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Itlay
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Asma Gul
- Department of Biological Sciences, International Islamic University Islamabad, Pakistan
| |
Collapse
|
11
|
Hiroki CH, Hassanabad MF, Defaye M, Sarden N, Bartlett A, Farias R, Nguyen AP, Guerrero-Fonseca IM, Yoon G, Brown L, Ma C, Yang H, Belke D, Hassanabad AF, McCoy C, Rosin NL, Orton DJ, Fedak PWM, Vallance BA, Yu H, Jacobson K, Khan N, Altier C, Kelly MM, Yipp BG. Nociceptor neurons suppress alveolar macrophage-induced Siglec-F + neutrophil-mediated inflammation to protect against pulmonary fibrosis. Immunity 2025:S1074-7613(25)00221-3. [PMID: 40449484 DOI: 10.1016/j.immuni.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/10/2025] [Accepted: 05/05/2025] [Indexed: 06/03/2025]
Abstract
Pulmonary fibrosis results from persistent and pathological tissue repair, which is therapeutically challenging to attenuate and often fatal. The immune processes involved in fibrosis remain ill defined. Using a bleomycin-induced lung fibrosis murine model, we discovered that vagal TRPV1+ nociceptors are protective. Pharmacological ablation or genetic deletion of nociceptors resulted in worsened fibrosis and outcomes. Without nociceptors, alveolar macrophages aberrantly produced vasoactive intestinal peptide (VIP), leading to cytokine TGF-β1-mediated alternative proinflammatory Siglec-F+ neutrophil recruitment to the lung with a high propensity for neutrophil extracellular trap (NET) formation. VIP inhibition or Vip deletion in hematopoietic cells improved outcomes and attenuated Siglec-F+ neutrophil recruitment to the lungs in nociceptor-deficient mice, while VIP administration had the opposite effect. Thus, nociceptors are essential regulators of inflammation during pulmonary fibrosis. These findings provide mechanistic insights into how the nervous system impacts the progression of fibrotic lung diseases.
Collapse
Affiliation(s)
- Carlos H Hiroki
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mortaza F Hassanabad
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Manon Defaye
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Nicole Sarden
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Alexandria Bartlett
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Raquel Farias
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Angela P Nguyen
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Idaira M Guerrero-Fonseca
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Grace Yoon
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Luke Brown
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Caixia Ma
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, Canada
| | - Hyungjun Yang
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, Canada
| | - Darrel Belke
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Christopher McCoy
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nicole L Rosin
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Dennis J Orton
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Bruce A Vallance
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, Canada
| | - Hongbing Yu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kevan Jacobson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, Canada
| | - Nargis Khan
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Christophe Altier
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Margaret M Kelly
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bryan G Yipp
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
12
|
Lee HL, Ju YH, Kim IY, Choi HJ, Heo YM, Na HR, Heo HJ. Codium fragile Extract Ameliorates Respiratory Function by Controlling Allergic Inflammation in Ovalbumin-Induced Bronchial Disorders in Mice. Mar Drugs 2025; 23:221. [PMID: 40422811 DOI: 10.3390/md23050221] [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: 04/17/2025] [Revised: 05/20/2025] [Accepted: 05/20/2025] [Indexed: 05/28/2025] Open
Abstract
This study investigated the effect of Codium fragile (WCF) water extract in reducing allergic inflammation in ovalbumin (OVA)-induced mice. Mice were sensitized to OVA + aluminum hydroxide, administered WCF for one week, and exposed to 1% aerosolized OVA. As a result, WCF intake reduced the OVA-induced increase in CD4+ T cells, CD8+ T cells, the T helper type 2 (Th2)/T helper type 1 (Th1) cell ratio, and inflammatory cells such as eosinophils and lymphocytes. Furthermore, WCF reduced Th2 cytokines such as interleukin (IL)-5, IL-13, and IL-33 and inflammatory cytokines such as tumor necrosis factor α (TNF-α) and IL-1β in lung tissues. A histological analysis showed that WCF intake decreases OVA-induced pulmonary inflammation, bronchial wall thickness, and mucus score and increases pulmonary alveolar area. Moreover, WCF inhibited the nuclear factor κB (NF-κB) pathway, the transforming growth factor β (TGF-β)/Smad pathway, and apoptosis-related proteins in lung tissues that OVA excessively activated. The oleamide (9-octadecenamide) content, representing a physiologically active component of WCF, was analyzed and validated using a high-performance liquid chromatography-photodiode array (HPLC-PDA) system. These results demonstrate that WCF may serve as a potential preventive agent for respiratory dysfunction such as allergic asthma by suppressing NF-κB and TGF-β/Smad pathways.
Collapse
Affiliation(s)
- Hyo Lim Lee
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Yeong Hyeon Ju
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - In Young Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hye Ji Choi
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Yu Mi Heo
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hwa Rang Na
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Ho Jin Heo
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
13
|
Chen X, Wu C, Tang F, Zhou J, Mo L, Li Y, He J. The Immune Microenvironment: New Therapeutic Implications in Organ Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05067. [PMID: 40391706 DOI: 10.1002/advs.202505067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/28/2025] [Indexed: 05/22/2025]
Abstract
Fibrosis, characterized by abnormal deposition of structural proteins, is a major cause of tissue dysfunction in chronic diseases. The disease burden associated with progressive fibrosis is substantial, and currently approved drugs are unable to effectively reverse it. Immune cells are increasingly recognized as crucial regulators in the pathological process of fibrosis by releasing effector molecules, such as cytokines, chemokines, extracellular vesicles, metabolites, proteases, or intercellular contact. Therefore, targeting the immune microenvironment can be a potential strategy for fibrosis reduction and reversion. This review summarizes the recent advances in the understanding of the immune microenvironment in fibrosis including phenotypic and functional transformations of immune cells and the interaction of immune cells with other cells. The novel opportunities for the discovery and development of drugs for immune microenvironment remodeling and their associated challenges are also discussed.
Collapse
Affiliation(s)
- Xiangqi Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Wu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fei Tang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingyue Zhou
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Mo
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
14
|
Yang B, Rutkowski N, Ruta A, Gray-Gaillard E, Maestas DR, Kelly SH, Krishnan K, Wu X, Wu S, Chen A, Mejías JC, Hooks JST, Vanderzee I, Mensah P, Celik N, Eric M, Abraham P, Tam A, Housseau F, Pardoll DM, Sears CL, Elisseeff JH. Murine gut microbiota dysbiosis via enteric infection modulates the foreign body response to a distal biomaterial implant. Proc Natl Acad Sci U S A 2025; 122:e2422169122. [PMID: 40354538 PMCID: PMC12107164 DOI: 10.1073/pnas.2422169122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
The gut microbiota influences systemic immunity and the function of distal tissues, including the brain, liver, skin, lung, and muscle. However, the role of the gut microbiota in the foreign body response and fibrosis is largely unexplored. To investigate this connection, we perturbed the homeostasis of the murine gut microbiota via infection with the pathogenic bacterial species enterotoxigenic Bacteroides fragilis (ETBF) and implanted particulate material (mean particle size <600 μm) of the synthetic polymer polycaprolactone (PCL) into a distal muscle injury. ETBF infection in mice led to increased neutrophil and γδ T cell infiltration into the PCL implant site. ETBF infection alone promoted systemic inflammation, increased levels of neutrophils in lymphoid tissues, and altered skeletal muscle gene expression. At the PCL implant site, we found significant changes in the transcriptome of sorted stromal cells between infected and control mice, including differences related to ECM components such as proteoglycans and glycosaminoglycans. However, we did not observe ETBF-induced differences in fibrosis levels. These results demonstrate the ability of the gut microbiota to mediate long-distance effects such as immune and stromal responses to a distal biomaterial implant.
Collapse
Affiliation(s)
- Brenda Yang
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Natalie Rutkowski
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Anna Ruta
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Elise Gray-Gaillard
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - David R. Maestas
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Sean H. Kelly
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Kavita Krishnan
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Xinqun Wu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Shaoguang Wu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Allen Chen
- Department of Biomedical Engineering, Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD21218
| | - Joscelyn C. Mejías
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Joshua S. T. Hooks
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Isabel Vanderzee
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Patricia Mensah
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Nazmiye Celik
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Marie Eric
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Peter Abraham
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
| | - Ada Tam
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Franck Housseau
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Drew M. Pardoll
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Cynthia L. Sears
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21287
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD21287
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD21287
| | - Jennifer H. Elisseeff
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21231
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD21287
| |
Collapse
|
15
|
Van Acker N, Frenois FX, Gravelle P, Tosolini M, Syrykh C, Laurent C, Brousset P. Spatial mapping of innate lymphoid cells in human lymphoid tissues and lymphoma at single-cell resolution. Nat Commun 2025; 16:4545. [PMID: 40374674 PMCID: PMC12081901 DOI: 10.1038/s41467-025-59811-1] [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: 09/11/2024] [Accepted: 05/06/2025] [Indexed: 05/17/2025] Open
Abstract
Innate lymphoid cells (ILC) distribution and compartmentalization in human lymphoid tissues are incompletely described. Through combined multiplex immunofluorescence, multispectral imaging, and advanced computer vision methods, we provide a map of ILCs at the whole-slide single-cell resolution level, and study their proximity to T helper (Th) cells. The results show that ILC2 predominates in thymic medulla; by contrast, immature Th cells prevail in the cortex. Unexpectedly, we find that Th2-like and Th17-like phenotypes appear before complete T cell receptor gene rearrangements in these immature thymocytes. In the periphery, ILC2 are more abundant in lymph nodes and tonsils, penetrating lymphoid follicles. NK cells are uncommon in lymphoid tissues but abundant in the spleen, whereas ILC1 and ILC3 predominate in the ileum and appendix. Under pathogenic conditions, a deep perturbation of both ILC and Th populations is seen in follicular lymphoma compared with non-neoplastic conditions. Lastly, all ILCs are preferentially in close proximity to their Th counterparts. In summary, our histopathology tool help present a spatial mapping of human ILCs and Th cells, in normal and neoplastic lymphoid tissues.
Collapse
Affiliation(s)
- Nathalie Van Acker
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - François-Xavier Frenois
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Pauline Gravelle
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Marie Tosolini
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Charlotte Syrykh
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Camille Laurent
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Pierre Brousset
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France.
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France.
| |
Collapse
|
16
|
Pellicer-Garcia L, Molimard C, Mauvais O, Pernet D, Bonniaud P, Valnet-Rabier MB, Barnig C. Organizing pneumonia in patients on dupilumab therapy for nasal polyposis: A report of three cases. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2025:S2213-2198(25)00421-0. [PMID: 40345325 DOI: 10.1016/j.jaip.2025.04.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/15/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Affiliation(s)
| | - Chloé Molimard
- Department of Pathology, University Hospital of Besançon, Besançon, France
| | - Olivier Mauvais
- Department of Head and Neck, University Hospital of Besançon, France
| | - Didier Pernet
- Department of Chest Disease, University Hospital Besançon, Besançon, France
| | - Philippe Bonniaud
- Institut Universitaire du Poumon, Centre de Référence Constitutif des Maladies Pulmonaires Rares de l'Adultes de Dijon, Réseau OrphaLung, Filière RespiFil, Centre Hospitalier Universitaire Dijon-Bourgogne, Dijon, France; Inserm U1231, Equipe HSP-pathies, Université Bourgogne Europe, Dijon, France; CRISALIS/F-CRIN INSERM Network, France
| | - Marie-Blanche Valnet-Rabier
- Regional Pharmacovigilance Centre Franche Comté, Clinical Pharmacology Department, University Hospital Besançon, Besançon, France
| | - Cindy Barnig
- Department of Chest Disease, University Hospital Besançon, Besançon, France; CRISALIS/F-CRIN INSERM Network, France; Université Marie et Louis Pasteur, CHU Besançon, EFS, INSERM, UMR RIGHT, Besançon, France.
| |
Collapse
|
17
|
Qiu X, Wu J, Chen Z, Zhang Y, Cao L, Wang N, Teng J, Su C, Cheng C, Wang F, Chen W. Circulating inflammatory cytokines and risk of aortic stenosis: A Mendelian randomization analysis. Cytokine 2025; 189:156887. [PMID: 39986193 DOI: 10.1016/j.cyto.2025.156887] [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: 12/08/2023] [Revised: 09/18/2024] [Accepted: 02/05/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Observational studies have consistently reported positive associations between inflammatory biomarkers and the risk of developing aortic stenosis (AS). However, it is crucial to acknowledge that conventional observational studies are prone to various forms of bias, including reverse causation and residual confounding. To delve deeper into unraveling the potential causal relationship between inflammatory biomarkers and aortic stenosis, we conducted a comprehensive two-sample Mendelian randomization (MR) analysis. METHODS In order to explore the causal effect of exposure to various circulating cytokines on the risk of developing AS, we carefully selected AS datasets as the exposures from the summary statistics of the genome-wide association study (GWAS) conducted by FinnGen. The dataset consisted of a sample size of 3283 for AS cases and 210,463 for controls. To estimate the MR analysis, we primarily adopted the inverse variance weighted (IVW) method. Additionally, we employed complementary methods, including Weighted Median, MR Egger, Weighted Mode, and Simple Mode, to analyze the causal associations comprehensively. In order to assess the presence of heterogeneity, we utilized Cochran's Q statistic and MR-Egger regression. To ensure the robustness and consistency of our findings, we conducted a leave-one-out analysis. RESULT We observed a positive association between interleukin-18 (IL-18) levels and AS (odds ratio [OR] per standard deviation [SD] = 1.080; 95 % confidence interval [CI] 1.024 to 1.139), as well as between interferon-gamma levels (IFN-γ) and AS (OR per SD = 1.157; 95 % CI 1.028 to 1.302). Conversely, we found an inverse association between interleukin-13 (IL-13) levels and AS (OR per SD = 0.942; 95 % CI 0.890 to 0.997), as well as between interleukin-5 (IL-5) levels and AS (OR per SD = 0.892; 95 % CI 0.804 to 0.990). CONCLUSION Our research enhances the current understanding of the role of specific inflammatory biomarker pathways in aortic stenosis. Nevertheless, further validation is required to assess the viability of targeting these cytokines through pharmacological or lifestyle interventions as potential treatments for aortic stenosis.
Collapse
Affiliation(s)
- Xiaohan Qiu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Jijun Wu
- Department of interventional radiology, Zhongshan Torch Development Zone People's Hospital, Zhongshan City, Guangdong Province, China; Third Clinical School, Guangzhou Medical University, Guangzhou, China
| | - Zehao Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Yu Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Luying Cao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Ning Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Junlin Teng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Cong Su
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Congyi Cheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China
| | - Fen Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China.
| | - Wenqiang Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012 Jinan, China.
| |
Collapse
|
18
|
Janto NV, Gleizes AR, Sun SJ, Ari G, Rao V, Gracz AD. Tritrichomonas muris sensitizes the intestinal epithelium to doxorubicin-induced apoptosis. Am J Physiol Gastrointest Liver Physiol 2025; 328:G594-G609. [PMID: 40243204 DOI: 10.1152/ajpgi.00242.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/26/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
Doxorubicin (DXR) is a widely used chemotherapy drug that can induce severe intestinal mucositis. Although the influence of gut bacteria on DXR-induced damage has been documented, the role of eukaryotic commensals remains unexplored. We discovered Tritrichomonas muris (Tmu) in one of our mouse colonies exhibiting abnormal tuft cell hyperplasia, prompting an investigation into its impact on DXR-induced intestinal injury. Mice from Tmu-colonized and Tmu-excluded facilities were injected with DXR. Tissue morphology and gene expression were evaluated at acute injury (6 h) and regenerative (72 h and 120 h) phases. Changes to crypt and villus morphology were more subtle than previously reported and region-specific, with significantly shorter jejunal villi in Tmu+ mice at 72 h post-DXR compared with Tmu- controls. Most notably, we observed elevated rates of DXR-induced apoptosis, measured by cleaved caspase 3 (CC3) staining, in Tmu+ intestinal crypts at 6 h post-DXR. Tmu+ mice also exhibited reduced expression of active intestinal stem cell (aISC) marker Lgr5 and facultative ISC (fISC) marker Ly6a at 6 h post-DXR compared with Tmu- controls. Tmu, but not DXR, was associated with increased inflammation and expression of type 2 cytokines IL-5 and IL-13. Tmu+ mice also exhibited a decreased fecal abundance of Lactobacillus, which promotes gut barrier integrity, and reduced claudin expression, indicating potential barrier dysfunction that could explain the increase in DXR-induced apoptosis. These findings highlight the significant influence of commensal microbiota, particularly eukaryotic organisms like Tmu, on intestinal biology and response to chemotherapy, underscoring the complexity of gut microbiota interactions in drug-induced mucositis.NEW & NOTEWORTHY Our study found that the eukaryotic commensal Tritrichomonas muris (Tmu) significantly increases DXR-induced intestinal apoptosis in mice. Tmu also reduces Lgr5 expression post-DXR injury and elevates inflammation and type 2 cytokine expression in the absence of injury. 16S sequencing identifies decreased abundance of protective Lactobacillus in Tmu colonized mice, as well as decreased expression of barrier-forming claudins, which may explain increased apoptosis. These findings emphasize the complex role of microbiota in drug-induced intestinal damage.
Collapse
Affiliation(s)
- Nicolas V Janto
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, Georgia, United States
| | - Antoine R Gleizes
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Siyang J Sun
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Gurel Ari
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Vivek Rao
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Adam D Gracz
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, Georgia, United States
- Department of Human Genetics, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
19
|
Mansouri M, Imenshahidi M, Rameshrad M, Hosseinzadeh H. Effects of Tinospora cordifolia (giloy) on metabolic syndrome components: a mechanistic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4979-5009. [PMID: 39731594 DOI: 10.1007/s00210-024-03642-2] [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: 09/10/2024] [Accepted: 11/15/2024] [Indexed: 12/30/2024]
Abstract
Metabolic syndrome is a cluster of some conditions such as high blood sugar, high blood triglycerides, low HDL cholesterol, abdominal obesity, and high blood pressure. Introducing a drug or a food that manages the majority of these medical conditions is invaluable. Tinospora cordifolia, known as guduchi and giloy, is a medicinal herb in ayurvedic medicine that is used in the treatment of various diseased conditions and also as a food for the maintenance of health. Here, we reviewed the current evidence supporting the role of giloy in the development and treatment of metabolic syndrome components. Appropriate articles that have been published until May 2024 were carefully extracted from PubMed, Scopus, and WOS databases to write a narrative review systematically. Gathered data showed the beneficial effects of giloy on metabolic syndrome components: hyperlipidemia, obesity, atherosclerosis, hypertension, and especially diabetes mellitus. As diabetes and insulin resistance seem to be a central feature of metabolic syndrome and in turn, can cause dyslipidemia, obesity, and, atherosclerosis, these beneficial effects are predictable with the anti-diabetogenic property of giloy. In this review, the main mechanisms of action of giloy in metabolic syndrome components are discussed. Based on the results, although giloy has been less investigated, considerable studies provide evidence of its beneficial effects on different components of metabolic syndrome. Relevant clinical trials are necessary to validate the mentioned effects, safety, and optimum dose of this herbal medicine and its components in managing different components of metabolic syndrome and transition from bench to bedside.
Collapse
Affiliation(s)
- Mehran Mansouri
- Student Research Committee, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Imenshahidi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rameshrad
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Dos Santos ACC, Figueiredo-Vanzan D, Bentes J, Motta JM, Mata-Santos HA, Pyrrho ADS, Castelo-Branco MTL. Tetrylpyamethrazine alleviates hepatic fibrosis induced by experimental mansonic schistosomiasis. Inflammopharmacology 2025; 33:2833-2847. [PMID: 40268854 DOI: 10.1007/s10787-025-01759-1] [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: 01/14/2025] [Accepted: 04/11/2025] [Indexed: 04/25/2025]
Abstract
Hepatic fibrosis resulting from human mansonic schistosomiasis significantly impairs liver function and contributes substantially to morbidity associated with helminth infections. This pathological state develops following the deposition of helminth eggs within hepatic tissues, triggering a granulomatous inflammatory reaction. Schistosomiasis, a neglected tropical disease affecting approximately 240 million individuals globally, represents a major public health challenge. Although praziquantel (PZQ) is recommended by the World Health Organization (WHO) as the primary treatment for helminth infections, additional therapies are required to address the associated liver fibrosis. This study investigated the efficacy of tetramethylpyrazine (TMP), a natural compound known for its anti-inflammatory, antifibrotic, and hepatoprotective properties in various experimental models, in mitigating hepatic fibrosis induced by mansonic schistosomiasis. Our in vivo experiments demonstrated that TMP treatment significantly reduced hepatic granuloma size, as evidenced by histological analysis. Furthermore, our in vitro studies showed that TMP increased levels of the anti-inflammatory cytokine IL-10 while decreasing levels of the profibrotic cytokine IL-13 in a concentration-dependent manner. Immunofluorescence analysis also revealed that TMP effectively inhibited collagen deposition. Collectively, these findings suggest that TMP exhibits potential as an anti-inflammatory and antifibrotic agent for hepatic fibrosis resulting from Schistosoma mansoni infection.
Collapse
Affiliation(s)
- Ana Carolina Campos Dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Josiane Bentes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Maria Motta
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Morgana Teixeira Lima Castelo-Branco
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
21
|
Kratchmarov R, Dharia T, Buchheit K. Clinical efficacy and mechanisms of biologics for chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2025; 155:1401-1410. [PMID: 40132672 PMCID: PMC12058411 DOI: 10.1016/j.jaci.2025.03.011] [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: 01/01/2025] [Revised: 02/23/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
The management of chronic rhinosinusitis with nasal polyps (CRSwNP) can be challenging, particularly when standard treatments including intranasal corticosteroids and endoscopic sinus surgery do not result in adequate symptom control. CRSwNP is frequently characterized by a type 2 immune signature, and many patients have other comorbid type 2 conditions, including asthma. There are currently 3 biologic therapies approved for the treatment of CRSwNP-omalizumab, mepolizumab, and dupilumab-and there are promising therapies in development. Biologic therapies allow for improved patient quality of life in CRSwNP, reduction in need for systemic corticosteroid treatment and endoscopic sinus surgery, and improvement in treatment of comorbidities. Translational studies assessing how biologic therapies can modify inflammation in CRSwNP have allowed for a greater understanding of CRSwNP pathogenesis. We review CRSwNP clinical trial and real-world data on the effectiveness and safety of biologics, discuss their therapeutic mechanisms, assess outcomes of biologic therapy versus endoscopic sinus surgery, and discuss therapies in development and future directions.
Collapse
Affiliation(s)
- Radomir Kratchmarov
- Division of Allergy and Clinical Immunology, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, and the Jeff and Penny Vinik Center, Boston, Mass
| | - Tiffany Dharia
- Division of Allergy and Clinical Immunology, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, and the Jeff and Penny Vinik Center, Boston, Mass
| | - Kathleen Buchheit
- Division of Allergy and Clinical Immunology, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, and the Jeff and Penny Vinik Center, Boston, Mass.
| |
Collapse
|
22
|
Shang K, Qi X, Tian T, Shi H, Zhu Y, Zhang F. Regulation of the tuft cell-ILC2 circuit in intestinal mucosal immunity. Front Immunol 2025; 16:1568062. [PMID: 40356895 PMCID: PMC12066627 DOI: 10.3389/fimmu.2025.1568062] [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: 01/28/2025] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
The intestinal mucosal immune system maintains homeostasis through complex interactions between epithelial cells and innate lymphoid cells in the lamina propria. Tuft cells, previously overlooked intestinal epithelial cell types, detect parasites and metabolites via Sucnr1 and TAS2R receptors. They secrete IL-25, which activates type 2 innate lymphoid cell (ILC2) via the IL-25R receptor. ILC2 releases IL-13, resulting in further promotion of tuft and goblet cells from stem cells. This positive feedback loop amplifies the local type 2 immune response, combating parasitic infections. Tuft cells also recognize viruses and bacteria, but the role played by the tuft cell-ILC2 circuit in this process is not yet clear. Furthermore, tuft cell-ILC2 circuit is influenced by dietary fiber, intestinal microbiota, and other factors, contributing to new functions in maintaining intestinal homeostasis. In inflammatory bowel disease, this immunological circuit may be protective. This review summarizes the current understanding of the tuft cell-ILC2 circuit, its regulatory mechanisms, and potential implications in intestinal disease.Graphical abstract (by Figdraw 2.0).
Collapse
Affiliation(s)
- Kaiyu Shang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xinxin Qi
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tingting Tian
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Huidong Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yuejie Zhu
- Reproductive Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Fengbo Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
23
|
Su Q, Wang J, Huangfu Y, Gao R, Kong P, Gao Y, Song H, Zhang J, Huang P, Zhang C, Feng Z, Kong D, Wang W. An Off-the-Shelf Artificial Proregenerative Macrophage for Pressure Ulcer Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415886. [PMID: 40271715 DOI: 10.1002/advs.202415886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/27/2025] [Indexed: 04/25/2025]
Abstract
Cell therapy is a promising approach in regenerative medicine. However, maintaining the survival and function of injected or implanted therapeutic cells remains a substantial challenge to success. In vivo modulatory strategy for cell therapeutics has been recently developed, but suffers from limited regenerative efficacy in injured tissue microenvironment with chronic inflammation. Here, an off-the-shelf artificial macrophage (artM) assembled by M2 macrophages-derived lysate proteins-loaded poly (lactic-co-glycolic acid) (PLGA) microspheres coated by macrophage cell membrane is developed. The synthetic artM fabricated in batches maintains its bioactivity with long-term cryostorage. Significantly, artM recapitulates the essential inflammation-regulatory and proregenerative characteristics of endogenous macrophages, including initiating M2 macrophage polarization, resolving excessive inflammation by releasing anti-inflammatory cytokines and growth factors, neutralizing endotoxins and proinflammatory cytokines, augmenting T-helper 2 (TH2) immune response, and coordinating cell migration and proliferation. In mouse model of deep tissue pressure injury (DTPI), the artM induces tissue regeneration by modulating the inflammatory microenvironment, promoting angiogenesis, reducing scar deposition, and accelerating the renewal of skin appendages. Depletion of macrophages in mice with skin ulcers highlights the immunomodulatory and proangiogenic functions of artM as effective as autogenous macrophages. Collectively, the engineered artM represents a cell-free, proreparative alternative to immune cell therapy in chronic wound management.
Collapse
Affiliation(s)
- Qi Su
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Jingrong Wang
- Beijing Life Science Academy, Beijing, 102200, China
| | - Yini Huangfu
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Rui Gao
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Pengxu Kong
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Yu Gao
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Huijuan Song
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Ju Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Pingsheng Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Chuangnian Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Zujian Feng
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Weiwei Wang
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| |
Collapse
|
24
|
Peng Y, Sheng J, Liu T, He R, Xu P. Identifying key targets and immune environment in wound healing based on iron overload-related genes. Arch Dermatol Res 2025; 317:719. [PMID: 40252113 DOI: 10.1007/s00403-025-04140-y] [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: 11/08/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 04/21/2025]
Abstract
Wound healing (WH) poses a significant socio-economic burden due to its high incidence and recurrence rates. Iron overload (IO) could be a factor leading to delayed WH. This study thus analyzed IO-related genes (IORGs) in WH, offering possibilities for developing new therapeutic strategies. Differential gene expression (DEGs) analysis was conducted between the WH group and intact skin (IS) group, intersected with IORGs to obtain differentially expressed IORGs (DE-IORGs). Functional enrichment analysis and potential drug screening were performed on DE-IORGs. A protein-protein interaction (PPI) network of DE-IORGs was constructed, and hub genes were identified using CytoHubba and MCODE methods. ROC curves of hub genes were plotted, and their expression levels in WH and IS groups as well as inter-gene correlations were analyzed. Additionally, immune infiltration variances in WH and IS groups, along with miRNA and TFs of hub genes, were examined. Finally, the effect of EGFR on skin wound healing was verified by scratch healing assay. 39 DE-IORGs were predominantly enriched in signaling pathways like HIF-1 signaling pathway and Th17 cell differentiation. Potential drugs for treating WH (e.g., felbamate, SA-94315, GANT-58, rucaparib) were identified. Three hub genes related to IO in WH were pinpointed (HIF1A, CDKN2A, EGFR) with diagnostic value. Immune infiltration analysis showed higher levels of immune cells like endothelial cells and macrophages in the WH group. Additionally, 55 miRNAs (e.g., hsa-mir-200a-3p, hsa-mir-218-5p) and 2 TFs (L3MBTL2, ZNF76) regulating the three hub genes were predicted. Cell experiments showed that EGFR could promote skin wound healing. The study suggested HIF1A, CDKN2A, and EGFR as potential diagnostic biomarkers for effective WH diagnosis, offering new insights into identifying potenti1al therapeutic targets for WH treatment.
Collapse
Affiliation(s)
- Yinbo Peng
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Institute of Traumatic Medicine of Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201900, China
| | - Juxiang Sheng
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
| | - Tiantian Liu
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Institute of Traumatic Medicine of Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201900, China
| | - Ruizhe He
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Institute of Traumatic Medicine of Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201900, China
| | - Peng Xu
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China.
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China.
- Institute of Traumatic Medicine of Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201900, China.
| |
Collapse
|
25
|
Liu X, Luo A, Yang M, Luo J, Li H, Chen X, Mao B, Jiang H, Liu W. Baicalin restores innate lymphoid immune imbalance during exacerbation of COPD. Immunol Res 2025; 73:71. [PMID: 40234295 PMCID: PMC12000166 DOI: 10.1007/s12026-025-09629-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 04/10/2025] [Indexed: 04/17/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by immune dysregulation, including altered innate lymphoid cell (ILC) immune responses, particularly during exacerbations (ECOPD). Baicalin, a natural compound prevalent in various herbal medicines, has shown promise as a therapeutic candidate in ECOPD. However, its potential and molecular mechanism for addressing ILC immune imbalance during ECOPD remain poorly understood. First, this study conducted a cross-sectional analysis of ILC immune responses in stable COPD patients and those experiencing exacerbations. Then, clinical findings of skewed ILC immunity were validated in cigarette smoke and lipopolysaccharide-induced ECOPD mouse models. Lastly, the therapeutic effect of baicalin on restoring ILC immune homeostasis was investigated in experimental ECOPD mouse models. Significant downregulation of ILC2 immunity was observed during COPD exacerbations, accompanied by increased ILC1 and ILC3 responses, particularly in cases associated with bacterial infections. Notably, elevated IL-22 levels were observed in this group. Administration of recombinant IL-22 in ECOPD mouse models disrupted lung ILC homeostasis, specifically inhibiting the accumulation of ILC2. Proteomics and transcriptomics analyses suggested IL-22 as a mediator of type 2 immune suppression by creating a molecular environment that favors type 1 and type 3 immunity. Treatment with baicalin effectively restored ILC2 immunity by enhancing the recruitment and activation of lung ILC2 while suppressing ILC1 and ILC3 responses. Importantly, baicalin attenuated IL-22 production from lung ILC3, highlighting its potential as an IL-22 inhibitor. Baicalin demonstrates potential as a therapeutic strategy for addressing ILC immune imbalance in COPD exacerbations, particularly by restoring ILC2 immunity and partially inhibiting IL-22 production. Clinical registration The cross-sectional study was registered with the Chinese Clinical Trial Registry (ChiCTR2100050683).
Collapse
Affiliation(s)
- Xuemei Liu
- Department of Internal Medicine, Division of Pulmonary Medicine, Institute of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Ai Luo
- Department of Internal Medicine, Division of Pulmonary Medicine, Institute of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Mei Yang
- Department of Internal Medicine, Division of Pulmonary Medicine, Institute of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
- Department of Pulmonary Medicine, Dazhou Second People's Hospital, Dazhou, China
| | - Jian Luo
- Respiratory Medicine Unit and National Institute for Health Research (NIHR), Nuffield Department of Medicine Experimental Medicine, Oxford Biomedical Research Centre (BRC), University of Oxford, Oxfordshire, United Kingdom
| | - Huifang Li
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, 1 Keyuansi Road, Chengdu, Sichuan, China
| | - Bing Mao
- Department of Internal Medicine, Division of Pulmonary Medicine, Institute of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hongli Jiang
- Department of Internal Medicine, Division of Pulmonary Medicine, Institute of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Wei Liu
- Department of Internal Medicine, Division of Pulmonary Medicine, Institute of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
26
|
Rembe JD, Garabet W, Augustin M, Dissemond J, Ibing W, Schelzig H, Stuermer EK. Immunomarker profiling in human chronic wound swabs reveals IL-1 beta/IL-1RA and CXCL8/CXCL10 ratios as potential biomarkers for wound healing, infection status and regenerative stage. J Transl Med 2025; 23:407. [PMID: 40200385 PMCID: PMC11978031 DOI: 10.1186/s12967-025-06417-2] [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: 08/30/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Chronic wounds, such as diabetic foot ulcers, venous leg ulcers, and post-surgical wound healing disorders pose a significant challenge due to prolonged healing, risk of infection, and impaired quality of life. Persistent inflammation and impaired tissue remodeling are common in these wounds. Traditional diagnostic methods, including visual inspection and microbiological cultures, offer limited insight into the wound micro-environment. Immunomarker profiling could provide a deeper understanding of the molecular mechanisms underpinning wound healing, offering potential biomarkers for infection status and healing progression. METHODS This observational, multi-center cohort study, part of the 'Wound-BIOME' project, analyzed 110 swab samples from patients with acute and chronic wounds using multiplex immunoassays. Clinical parameters such as wound type, healing status, regeneration stage, and microbial burden were recorded. Total protein concentration was assessed, and 35 key immunomarkers, including cytokines (e.g. IL- 1α, IL- 1β), chemokines (CCL2, CXCL8, CXCL10), growth factors (FGF- 2, VEGF) and matrix metalloproteinases (MMP- 7, MMP- 9, MMP- 13), were quantified. Statistical analyses were performed to correlate immunomarker levels with clinical outcomes. RESULTS Pro-inflammatory markers, such as IL- 1β, IL- 18 and chemokines like CCL2 and CXCL8, were significantly elevated in non-healing and infected wounds compared to healing wounds. The study identified two new immunomarker ratios - IL- 1β/IL- 1RA and CXCL8/CXCL10 - as potential predictors of wound healing status. The IL- 1β/IL- 1RA ratio showed the highest accuracy for distinguishing healing from non-healing wounds (AUC = 0.6837), while the CXCL8/CXCL10 ratio was most effective in identifying infection (AUC = 0.7669). CONCLUSIONS Immunomarker profiling via wound swabbing offers valuable insights into the wound healing process. Elevated levels of pro-inflammatory cytokines and MMPs are associated with chronic inflammation and impaired healing. The IL- 1β/IL- 1RA and CXCL8/CXCL10 ratios emerge as promising biomarkers to distinguish between infection and inflammation, with potential in targeted wound care. Further studies are needed to validate these findings and implement them in clinical practice.
Collapse
Affiliation(s)
- Julian-Dario Rembe
- Department for Vascular and Endovascular Surgery, University Hospital Duesseldorf (UKD), Heinrich Heine University Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany.
| | - Waseem Garabet
- Department for Vascular and Endovascular Surgery, University Hospital Duesseldorf (UKD), Heinrich Heine University Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany
| | - Matthias Augustin
- Institute for Health Services Research in Dermatology and Nursing Professions (IVDP), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Joachim Dissemond
- Department of Dermatology, Venereology and Allergology, Essen University Hospital, Essen, Germany
| | - Wiebke Ibing
- Department for Vascular and Endovascular Surgery, University Hospital Duesseldorf (UKD), Heinrich Heine University Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany
| | - Hubert Schelzig
- Department for Vascular and Endovascular Surgery, University Hospital Duesseldorf (UKD), Heinrich Heine University Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany
| | - Ewa K Stuermer
- Clinic and Polyclinic for Vascular Medicine, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
27
|
Roberts LB, Kelly AM, Hepworth MR. There's no place like home: How local tissue microenvironments shape the function of innate lymphoid cells. Mucosal Immunol 2025; 18:279-289. [PMID: 39900201 DOI: 10.1016/j.mucimm.2025.01.012] [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: 11/21/2024] [Revised: 01/16/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
Innate lymphoid cells (ILC) have emerged as critical immune effectors with key roles in orchestrating the wider immune response. While ILC are relatively rare cells they are found enriched within discrete microenvironments, predominantly within barrier tissues. An emerging body of evidence implicates complex and multi-layered interactions between cell types, tissue structure and the external environment as key determinants of ILC function within these niches. In this review we will discuss the specific components that constitute ILC-associated microenvironments and consider how they act to determine health and disease. The development of holistic, integrated models of ILC function within complex tissue environments will inform new understanding of the contextual cues and mechanisms that determine the protective versus disease-causing roles of this immune cell family.
Collapse
Affiliation(s)
- Luke B Roberts
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester M13 9PL United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, United Kingdom
| | - Alanna M Kelly
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester M13 9PL United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, United Kingdom
| | - Matthew R Hepworth
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester M13 9PL United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, United Kingdom.
| |
Collapse
|
28
|
Wang J, Yin J, Liu X, Liu Y, Jin X. Gut commensal bacterium Bacteroides vulgatus exacerbates helminth-induced cardiac fibrosis through succinate accumulation. PLoS Pathog 2025; 21:e1013069. [PMID: 40238740 PMCID: PMC12002503 DOI: 10.1371/journal.ppat.1013069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/21/2025] [Indexed: 04/18/2025] Open
Abstract
Trichinella spiralis (Ts) is known to cause cardiac fibrosis, which is a critical precursor to various heart diseases, and its progression is influenced by metabolic changes. However, the metabolic mechanisms remain unclear. Here, we observed that Ts-infected mice exhibited cardiac fibrosis along with elevated succinate levels in the heart using metabolomic analysis. Administration of succinate exacerbated fibrosis during Ts infection, while deficiency in succinate receptor 1 (Sucnr1) alleviated the condition, highlighting the role of the succinate-Sucnr1 axis in fibrosis development. Furthermore, metagenomics sequencing showed that Ts-infected mice had a higher abundance ratio of succinate-producing bacteria to succinate-consuming bacteria in the intestines. Notably, the succinate-producer Bacteroides vulgatus was enriched in Ts group. Oral supplementation with B. vulgatus aggravated Ts-induced cardiac fibrosis. In summary, our findings underscore the succinate-Sucnr1 axis as a critical pathway in helminth-induced cardiac fibrosis and highlight the potential of targeting this axis for therapeutic interventions. This study presents novel insights into the gut-heart axis, revealing innovative strategies for managing cardiovascular complications associated with helminth infections.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
- College of Animal Sciences, Jilin University, Changchun, China
| | - Jiali Yin
- The Second Hospital of Jilin University, Changchun, China
| | - Xiaolei Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious 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 Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Xuemin Jin
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
29
|
Vasarmidi E, Worrell JC, Mahmutovic Persson I, Yaqub N, Miądlikowska E, Barnig C, Boots A, Reynaert NL, Cuevas Ocaña S. Insights into interstitial lung disease pathogenesis. Breathe (Sheff) 2025; 21:240261. [PMID: 40365095 PMCID: PMC12070197 DOI: 10.1183/20734735.0261-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/11/2025] [Indexed: 05/15/2025] Open
Abstract
This review summarises some of the key features of interstitial lung diseases (ILDs) from a translational science point of view and brings insights into potential therapeutic options. Genetic predisposition and environmental factors like smoking, pollution and infections significantly impact the onset, progression and treatment response in ILDs, highlighting the need for personalised management. Fibroblasts are central to ILD pathology, influencing the tissue microenvironment, immune cell interactions and extracellular matrix (ECM) production, making them critical therapeutic targets. Monocyte-derived M2 macrophages drive fibrosis in idiopathic pulmonary fibrosis by secreting cytokines and remodelling the ECM. Understanding macrophage subtypes and their dynamics offers new therapeutic possibilities. Chronic type 2 immunity contributes to fibrosis, emphasising the need to enhance protective markers in order to even out the balance shift of pathological immune responses in ILD treatments. Serum biomarkers like Krebs von den Lungen-6 (KL-6), surfactant protein (SFTP) D, matrix metalloproteinase-7 (MMP-7), and C-C motif chemokine ligand (CCL)-18 are valuable for diagnosing and predicting ILD progression, although more research is needed for clinical application. Animal models, especially bleomycin-based models, offer insights into ILD pathology, but challenges like lung hyperinflation highlight the need for careful model selection and translational research to bridge preclinical and clinical findings.
Collapse
Affiliation(s)
- Eirini Vasarmidi
- Department of Respiratory Medicine, Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, Heraklion, Greece
- These authors contributed equally
| | - Julie C. Worrell
- Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
- These authors contributed equally
| | - Irma Mahmutovic Persson
- Respiratory Immunopharmacology, Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
- Lund University BioImaging Centre (LBIC), Faculty of Medicine, Lund University, Lund, Sweden
- These authors contributed equally
| | - Naheem Yaqub
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Ewa Miądlikowska
- Department of Pneumology, Medical University of Lodz, Lodz, Poland
| | - Cindy Barnig
- Université de Franche-Comté, CHU Besançon, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Agnes Boots
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Niki L. Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Sara Cuevas Ocaña
- Biodiscovery Institute, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
30
|
Merugu S, Jagiello K, Gajewicz-Skretna A, Halappanavar S, Willliams A, Vogel U, Puzyn T. The Impact of Carbon Nanotube Properties on Lung Pathologies and Atherosclerosis Through Acute Inflammation: a New AOP-Anchored in Silico NAM. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2501185. [PMID: 40025979 DOI: 10.1002/smll.202501185] [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: 01/28/2025] [Indexed: 03/04/2025]
Abstract
In this study, a previously developed approach for creating a quantitative structure-activity relationship model anchored in an Adverse Outcome Pathway framework (AOP-anchored Nano-QSAR) is employed to develop a novel model capable of predicting transcriptomic responses triggered by the inhalation of multiwalled carbon nanotubes (MWCNTs). The acute phase response (AR) signaling pathway, which plays a crucial role in neutrophil influx and initiates the acute immune response is focused. This process involves recruiting pro-inflammatory cells into the lungs and can lead to lung fibrosis, as outlined in AOP33, or atherosclerosis, as per AOP237. To establish the relationship between the structural properties of a set of MWCNTs and the transcriptional benchmark dose level (BMDLAR) response of genes associated with the acute phase response signaling pathway, the locally weighted kernel linear regression algorithm is used. These findings emphasize the critical role of the aspect ratio and specific surface area of MWCNTs in initiating acute inflammation and, subsequently, lung pathologies and atherosclerosis through the inflammatory and acute phase response signaling pathways. This newly developed data-driven model extends the repertoire of transcriptomic-based, AOP-informed Nano-QSAR models, potentially serving as an in silico new approach methodology (NAM) to support the MWCNTs' safety assessment based on the weight of evidence.
Collapse
Affiliation(s)
- Sattibabu Merugu
- University of Gdansk, Faculty of Chemistry, Wita Stwosza 63, Gdansk, 80-308, Poland
| | - Karolina Jagiello
- University of Gdansk, Faculty of Chemistry, Wita Stwosza 63, Gdansk, 80-308, Poland
- QSAR Lab Ltd., Trzy lipy 3, Gdansk, 80-172, Poland
| | | | - Sabina Halappanavar
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, K1A 0K9, Canada
| | - Andrew Willliams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, K1A 0K9, Canada
| | - Ulla Vogel
- The National Research Centre for the Working Environment, Copenhagen, DK-2100, Denmark
| | - Tomasz Puzyn
- University of Gdansk, Faculty of Chemistry, Wita Stwosza 63, Gdansk, 80-308, Poland
- QSAR Lab Ltd., Trzy lipy 3, Gdansk, 80-172, Poland
| |
Collapse
|
31
|
Warrick KA, Vallez CN, Meibers HE, Pasare C. Bidirectional Communication Between the Innate and Adaptive Immune Systems. Annu Rev Immunol 2025; 43:489-514. [PMID: 40279312 PMCID: PMC12120936 DOI: 10.1146/annurev-immunol-083122-040624] [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] [Indexed: 04/27/2025]
Abstract
Effective bidirectional communication between the innate and adaptive immune systems is crucial for tissue homeostasis and protective immunity against infections. The innate immune system is responsible for the early sensing of and initial response to threats, including microbial ligands, toxins, and tissue damage. Pathogen-related information, detected primarily by the innate immune system via dendritic cells, is relayed to adaptive immune cells, leading to the priming and differentiation of naive T cells into effector and memory lineages. Memory T cells that persist long after pathogen clearance are integral for durable protective immunity. In addition to rapidly responding to reinfections, memory T cells also directly instruct the interacting myeloid cells to induce innate inflammation, which resembles microbial inflammation. As such, memory T cells act as newly emerging activators of the innate immune system and function independently of direct microbial recognition. While T cell-mediated activation of the innate immune system likely evolved as a protective mechanism to combat reinfections by virulent pathogens, the detrimental outcomes of this mechanism manifest in the forms of autoimmunity and other T cell-driven pathologies. Here, we review the complexities and layers of regulation at the interface between the innate and adaptive immune systems to highlight the implications of adaptive instruction of innate immunity in health and disease.
Collapse
Affiliation(s)
- Kathrynne A Warrick
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Charles N Vallez
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Hannah E Meibers
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| |
Collapse
|
32
|
Ye W, Meng X, Xu S. [Research progress on collagen secretion mechanisms in scarring]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2025; 54:266-278. [PMID: 40194913 PMCID: PMC12062945 DOI: 10.3724/zdxbyxb-2024-0535] [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: 09/24/2024] [Revised: 12/12/2024] [Accepted: 02/22/2025] [Indexed: 04/09/2025]
Abstract
Scar formation is characterized by dynamic alterations in collagen secretion, which critically determine scar morphology and pathological progression. In fibroblasts, collagen secretion is initiated through the activation of cytokine- and integrin-mediated signaling pathways, which promote collagen gene transcription. The procollagen polypeptide α chains undergo extensive post-translational modifications, including hydroxylation and glycosylation, within the endoplasmic reticulum (ER), followed by folding and assembly into triple-helical procollagen. Subsequent intracellular trafficking involves the sequential transport of procollagen through the ER, Golgi apparatus, and plasma membrane, accompanied by further structural refinements prior to extracellular secretion. Once secreted, procollagen is enzymatically processed to form mature collagen fibrils, which drive scar tissue remodeling. Recent advances in elucidating regulation of collagen secretion have identified pivotal molecular targets, such as transforming growth factor-beta 1 (TGF-β1), prolyl 4-hydroxylase (P4H), heat shock protein 47 (HSP47), and transport and Golgi organization protein 1 (TANGO1), providing novel therapeutic strategies to mitigate pathological scar hyperplasia and improve regenerative outcomes. This review provides a comprehensive analysis of the molecular mechanisms governing collagen secretion during scar formation, with emphasis on signaling cascades, procollagen biosynthesis, intracellular transport dynamics, and post-translational modifications, thereby offering a framework for developing targeted anti-scar therapies.
Collapse
Affiliation(s)
- Wenkai Ye
- Center of Stem Cell and Regenerative Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Xinan Meng
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, International Institutes of Medicine, Zhejiang University, Center for Membrane Receptors and Brain Medicine, International School of Medicine, Zhejiang University, Yiwu 322000, Zhejiang Province, China
| | - Suhong Xu
- Center of Stem Cell and Regenerative Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
33
|
Rieder F, Nagy LE, Maher TM, Distler JHW, Kramann R, Hinz B, Prunotto M. Fibrosis: cross-organ biology and pathways to development of innovative drugs. Nat Rev Drug Discov 2025:10.1038/s41573-025-01158-9. [PMID: 40102636 DOI: 10.1038/s41573-025-01158-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/20/2025]
Abstract
Fibrosis is a pathophysiological mechanism involved in chronic and progressive diseases that results in excessive tissue scarring. Diseases associated with fibrosis include metabolic dysfunction-associated steatohepatitis (MASH), inflammatory bowel diseases (IBDs), chronic kidney disease (CKD), idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc), which are collectively responsible for substantial morbidity and mortality. Although a few drugs with direct antifibrotic activity are approved for pulmonary fibrosis and considerable progress has been made in the understanding of mechanisms of fibrosis, translation of this knowledge into effective therapies continues to be limited and challenging. With the aim of assisting developers of novel antifibrotic drugs, this Review integrates viewpoints of biologists and physician-scientists on core pathways involved in fibrosis across organs, as well as on specific characteristics and approaches to assess therapeutic interventions for fibrotic diseases of the lung, gut, kidney, skin and liver. This discussion is used as a basis to propose strategies to improve the translation of potential antifibrotic therapies.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA.
- Program for Global Translational Inflammatory Bowel Diseases (GRID), Chicago, IL, USA.
| | - Laura E Nagy
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
| | - Toby M Maher
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- National Heart and Lung Institute, Imperial College, London, UK
| | - Jörg H W Distler
- Department of Rheumatology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen; Medical Faculty, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
34
|
Mandviwala AS, Liman K, Huckriede ALW, Arankalle VA, Patil HP. Evaluation of dual pathogen recognition receptor agonists as adjuvants for respiratory syncytial virus - virus-like particles for pulmonary delivery. Front Immunol 2025; 16:1561297. [PMID: 40176816 PMCID: PMC11962540 DOI: 10.3389/fimmu.2025.1561297] [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: 01/15/2025] [Accepted: 02/25/2025] [Indexed: 04/04/2025] Open
Abstract
Introduction Respiratory syncytial virus (RSV) remains a significant global health concern, particularly for infants and young children in developing countries. Despite ongoing research efforts, an effective RSV vaccine has yet to be approved for widespread use. Use of two separate pattern recognition receptor (PRR) agonists as adjuvants in vaccine formulations has shown to enhance the immune response against the antigen. The limitation with the use of two adjuvants is that they need not necessarily bind to PRRs on the same cell. This study evaluates the efficacy of two different dual PRR binding chimeric molecules CL413 (TLR2/TLR7 agonist) and CL429 (TLR2/NOD2 agonist) as adjuvants for RSV virus-like particles (VLPs) delivered via the pulmonary route in mice for induction of mucosal and systemic immunity. Methods BALB/c mice were immunized twice with the RSV-VLPs alone or adjuvanted with CL413, CL429, mixture of single PRR agonists Pam3CSK4+ L18-MDP or Pam3CSK4+ imiquimod via the pulmonary route. The mixture of single PRR agonists adjuvants was used as control for chimeric adjuvants. Immune responses were evaluated by measuring antibody levels in sera and respiratory tract; cytokine production, B and T cell responses in the lungs and spleen. Results Pulmonary immunization with CL413-adjuvanted VLPs induced robust nasal IgA responses against the RSV F and G proteins, which was not observed for the other adjuvant combinations. CL413 also enhanced serum IgG levels and promoted a balanced Th1/Th2 response, as evidenced by IgG2a/IgG1 ratios. CL413 elicited strong pro-inflammatory responses in the lungs of mice, including elevated levels of IFN-γ, TNF-α, IL-6, and IL-17A. Flow cytometry analysis revealed increased numbers of tissue-resident class-switched B cells in the lungs of mice that were immunized with VLPs adjuvanted with CL413 and CL429. CD4+ and CD8+ T cell responses were also enhanced in both lungs and spleens of mice receiving VLPs adjuvanted with chimeric molecules to various extents. Mice immunized with formalin inactivated RSV (FI-RSV), which are used as the positive control for vaccine induced pathology after RSV challenge developed alveolitis, perivascular infiltration. While all the mice receiving adjuvanted VLP formulations showed protection against lung pathology after RSV challenge. Discussion The lack of pathology, combined with the robust mucosal and systemic immune responses, suggests that pulmonary delivery of adjuvanted RSV-VLPs may provide effective protection without the risk of vaccine-enhanced disease. The study also demonstrates that the chimeric TLR2/TLR7 agonist CL413 is a promising adjuvant for RSV-VLPs to induce mucosal and systemic immune response and warrant further investigations in more advanced preclinical models.
Collapse
MESH Headings
- Animals
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Mice
- Mice, Inbred BALB C
- Respiratory Syncytial Virus Vaccines/immunology
- Respiratory Syncytial Virus Vaccines/administration & dosage
- Respiratory Syncytial Virus Infections/immunology
- Respiratory Syncytial Virus Infections/prevention & control
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Female
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Lung/immunology
- Receptors, Pattern Recognition/agonists
- Humans
- Respiratory Syncytial Virus, Human/immunology
- Cytokines
- Immunity, Mucosal
- Respiratory Syncytial Viruses/immunology
Collapse
Affiliation(s)
- Ahmedali S. Mandviwala
- Department of Translational Virology, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Komal Liman
- Department of Translational Virology, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Anke L. W. Huckriede
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Vidya A. Arankalle
- Department of Translational Virology, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Harshad P. Patil
- Department of Translational Virology, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| |
Collapse
|
35
|
Chen C, Zhen Z, Cui M, Hu X, Yu L, Zhou F, Yu X, Yang D, Wu H, Cui Y, Li X, Cui X, Liang X, Gao Y, Liu Y, Yu Y, Huang Z, Zhang F. The core cellular network modulates immune phenotype switching in hepatitis B. Sci Bull (Beijing) 2025:S2095-9273(25)00289-0. [PMID: 40175178 DOI: 10.1016/j.scib.2025.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Affiliation(s)
- Chuangeng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Ziqi Zhen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Meng Cui
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Xiaoli Hu
- Department of Infectious Diseases, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin 150030, China
| | - Lei Yu
- Department of Infectious Diseases, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Fengxia Zhou
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Xiaorong Yu
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Dehui Yang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - He Wu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, China
| | - Ying Cui
- Department of Infectious Diseases, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin 150030, China
| | - Xiang Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Xudong Cui
- Department of Infectious Diseases, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xinyue Liang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Yiyang Gao
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yuchen Liu
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yang Yu
- Harbin Institute of Technology Hospital, Harbin 150006, China
| | - Zhiwei Huang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China.
| | - Fan Zhang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China.
| |
Collapse
|
36
|
de Zawadzki A, Leeming DJ, Sanyal AJ, Anstee QM, Schattenberg JM, Friedman SL, Schuppan D, Karsdal MA. Hot and cold fibrosis: The role of serum biomarkers to assess immune mechanisms and ECM-cell interactions in human fibrosis. J Hepatol 2025:S0168-8278(25)00148-5. [PMID: 40056933 DOI: 10.1016/j.jhep.2025.02.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 05/24/2025]
Abstract
Fibrosis is a pathological condition characterised by excessive accumulation of extracellular matrix (ECM) components, particularly collagens, leading to tissue scarring and organ dysfunction. In fibrosis, an imbalance between collagen synthesis (fibrogenesis) and degradation (fibrolysis) results in the deposition of fibrillar collagens disrupting the structural integrity of the ECM and, consequently, tissue architecture. Fibrosis is associated with a wide range of chronic diseases, including cirrhosis, kidney fibrosis, pulmonary fibrosis, and autoimmune diseases. Recently, the concept of "hot" and "cold" fibrosis has emerged, referring to the immune status within fibrotic tissues and the nature of fibrogenic signalling. Hot fibrosis is characterised by active immune cell infiltration and inflammation, while cold fibrosis is associated with auto- and paracrine myofibroblast activation, immune cell exclusion and quiescence. In this article, we explore the relationship between hot and cold fibrosis, the role of various types of collagens and their biologically active fragments in modulating the immune system, and how serological ECM biomarkers can help improve our understanding of the disease-relevant interactions between immune and mesenchymal cells in fibrotic tissues. Additionally, we draw lessons from immuno-oncology research in solid tumours to shed light on potential strategies for fibrosis treatment and highlight the advantage of having a "hot fibrotic environment" to treat fibrosis by enhancing collagen degradation through modulation of the immune system.
Collapse
Affiliation(s)
| | - Diana J Leeming
- Nordic Bioscience A/S, Biomarkers & Research, Herlev, Denmark
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health and Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; Newcastle NIHR Biomedical Research Center, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| | - Jörn M Schattenberg
- Department of Internal Medicine II, Saarland University Medical Centre, Homburg, Germany
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, Harvard Medical School, MA, USA
| | | |
Collapse
|
37
|
Bamias G, Menghini P, Pizarro TT, Cominelli F. Targeting TL1A and DR3: the new frontier of anti-cytokine therapy in IBD. Gut 2025; 74:652-668. [PMID: 39266053 PMCID: PMC11885054 DOI: 10.1136/gutjnl-2024-332504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/20/2024] [Indexed: 09/14/2024]
Abstract
TNF-like cytokine 1A (TL1A) and its functional receptor, death-domain receptor 3 (DR3), are members of the TNF and TNFR superfamilies, respectively, with recognised roles in regulating innate and adaptive immune responses; additional existence of a decoy receptor, DcR3, indicates a tightly regulated cytokine system. The significance of TL1A:DR3 signalling in the pathogenesis of inflammatory bowel disease (IBD) is supported by several converging lines of evidence. Herein, we aim to provide a comprehensive understanding of what is currently known regarding the TL1A/DR3 system in the context of IBD. TL1A and DR3 are expressed by cellular subsets with important roles for the initiation and maintenance of intestinal inflammation, serving as potent universal costimulators of effector immune responses, indicating their participation in the pathogenesis of IBD. Recent evidence also supports a homoeostatic role for TL1A:DR3 via regulation of Tregs and innate lymphoid cells. TL1A and DR3 are also expressed by stromal cells and may contribute to inflammation-induced or inflammation-independent intestinal fibrogenesis. Finally, discovery of genetic polymorphisms with functional consequences may allow for patient stratification, including differential responses to TL1A-targeted therapeutics. In conclusion, TL1A:DR3 signalling plays a central and multifaceted role in the immunological pathways that underlie intestinal inflammation, such as that observed in IBD. Such evidence provides the foundation for developing pharmaceutical approaches targeting this ligand-receptor pair in IBD.
Collapse
Affiliation(s)
- Giorgos Bamias
- GI Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Paola Menghini
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
38
|
Chen H, Zhou X, Liu T, Liu J, Wu D, Xu X, Ma S, Qiang G, Chen J, Cao Y, Fu W, Yang J. Postprandial parasympathetic signals promote lung type 2 immunity. Neuron 2025; 113:670-683.e7. [PMID: 39837323 DOI: 10.1016/j.neuron.2024.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 11/03/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025]
Abstract
Lung type 2 immunity protects against pathogenic infection, but its dysregulation causes asthma. Although it has long been observed that symptoms of asthmatic patients often become exaggerated following food intake, the pathophysiological mechanism underlying this postprandial phenomenon is incompletely understood. Here, we report that lung type 2 immunity in mice is enhanced after feeding, which correlates with parasympathetic activation. Also, local parasympathetic innervations exhibit spatial engagement with such immune responses mediated by group 2 innate lymphoid cells (ILC2s). Pharmacologic or surgical blockage of parasympathetic signals diminishes lung type 2 immunity. Conversely, chemogenetic manipulation of parasympathetic inputs and their upstream neurocircuit is sufficient to modulate those immune responses. We then show that the cholinergic receptor muscarinic 4 (Chrm4) for the parasympathetic neurotransmitter acetylcholine is expressed in mouse or human lung ILC2s, and the Chrm4 deletion mitigates ILC2-mediated lung inflammation. These results have revealed a critical neuroimmune function of the gut-brain-lung reflex.
Collapse
Affiliation(s)
- Hongjie Chen
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Xin Zhou
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; Department of General Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Tingting Liu
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Jiaqi Liu
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Di Wu
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Xia Xu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shanwu Ma
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Guangliang Qiang
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Jian Chen
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Ying Cao
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Wei Fu
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; Department of General Surgery, Peking University Third Hospital, Beijing 100191, China.
| | - Jing Yang
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; Peking Union Medical College Hospital, Beijing 100730, China.
| |
Collapse
|
39
|
Ziglari T, Calistri NL, Finan JM, Derrick DS, Nakayasu ES, Burnet MC, Kyle JE, Hoare M, Heiser LM, Pucci F. Senescent Cell-Derived Extracellular Vesicles Inhibit Cancer Recurrence by Coordinating Immune Surveillance. Cancer Res 2025; 85:859-874. [PMID: 39804967 PMCID: PMC11878441 DOI: 10.1158/0008-5472.can-24-0875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/28/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Senescence is a nonproliferative survival state that cancer cells can enter to escape therapy. In addition to soluble factors, senescence cells secrete extracellular vesicles (EV), which are important mediators of intercellular communication. To explore the role of senescent cell (SC)-derived EVs (senEV) in inflammatory responses to senescence, we developed an engraftment-based senescence model in wild-type mice and genetically blocked senEV release in vivo, without significantly affecting soluble mediators. SenEVs were both necessary and sufficient to trigger immune-mediated clearance of SCs, thereby suppressing tumor growth. In the absence of senEVs, the recruitment of MHC-II+ antigen-presenting cells (APC) to the senescence microenvironment was markedly impaired. Blocking senEV release redirected the primary target of SC signaling from APCs to neutrophils. Comprehensive transcriptional and proteomic analyses identified six ligands specific to senEVs, highlighting their role in promoting APC-T cell adhesion and synapse formation. APCs activated CCR2+CD4+ TH17 cells, which seemed to inhibit B-cell activation, and CD4+ T cells were essential for preventing tumor recurrence. These findings suggest that senEVs complement the activity of secreted inflammatory mediators by recruiting and activating distinct immune cell subsets, thereby enhancing the efficient clearance of SCs. These conclusions may have implications not only for tumor recurrence but also for understanding senescence during de novo carcinogenesis. Consequently, this work could inform the development of early detection strategies for cancer based on the biology of cellular senescence. Significance: Chemotherapy-treated senescent tumor cells release extracellular vesicles that trigger an immune response and suppress tumor recurrence. See related commentary by Almeida and Melo, p. 833.
Collapse
Affiliation(s)
- Tahereh Ziglari
- Department of Otolaryngology – Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon, US
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon, US
- Current address: Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, US
| | - Nicholas L. Calistri
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, US
| | - Jennifer M. Finan
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon, US
| | - Daniel S. Derrick
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, US
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, US
| | - Meagan C. Burnet
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, US
| | - Jennifer E. Kyle
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, US
| | - Matthew Hoare
- Early Cancer Institute, University of Cambridge, Cambridge, UK
| | - Laura M. Heiser
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, US
| | - Ferdinando Pucci
- Department of Otolaryngology – Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon, US
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon, US
| |
Collapse
|
40
|
Zhang S, Dong H, Jin X, Sun J, Li Y. The multifaceted roles of macrophages in the transition from hepatitis to hepatocellular carcinoma: From mechanisms to therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167676. [PMID: 39828046 DOI: 10.1016/j.bbadis.2025.167676] [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: 11/28/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Macrophages are central to the progression from hepatitis to hepatocellular carcinoma (HCC), with their remarkable plasticity and ability to adapt to the changing liver microenvironment. Chronic inflammation, fibrosis, and ultimately tumorigenesis are driven by macrophage activation, making them key regulators of liver disease progression. This review explores the diverse roles of macrophages in the transition from hepatitis to HCC. In the early stages of hepatitis, macrophages are essential for pathogen clearance and tissue repair. However, chronic activation leads to prolonged inflammation, which exacerbates liver damage and promotes fibrosis. As the disease progresses to liver fibrosis, macrophages interact with hepatic stellate cells, fostering a pro-tumorigenic microenvironment that supports HCC development. In hepatocarcinogenesis, macrophages contribute to tumor initiation, growth, metastasis, immune evasion, cancer stem cell maintenance, and angiogenesis. Their functional plasticity enables them to adapt to the tumor microenvironment, thereby promoting tumor progression and resistance to therapy. Targeting macrophages represents a promising strategy for preventing and treating HCC. Therapeutic approaches, including reprogramming macrophage phenotypes to enhance anti-tumor immunity, blocking macrophage recruitment and activation, and utilizing nanoparticle-based drug delivery systems, may provide new avenues for combating HCC by modulating macrophage functions and tumor microenvironment dynamics.
Collapse
Affiliation(s)
- Shuairan Zhang
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Hang Dong
- Phase I Clinical Trials Center, The People's Hospital of China Medical University, Shenyang, PR China
| | - Xiuli Jin
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Jing Sun
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Yiling Li
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
41
|
Ogulur I, Mitamura Y, Yazici D, Pat Y, Ardicli S, Li M, D'Avino P, Beha C, Babayev H, Zhao B, Zeyneloglu C, Giannelli Viscardi O, Ardicli O, Kiykim A, Garcia-Sanchez A, Lopez JF, Shi LL, Yang M, Schneider SR, Skolnick S, Dhir R, Radzikowska U, Kulkarni AJ, Imam MB, Veen WVD, Sokolowska M, Martin-Fontecha M, Palomares O, Nadeau KC, Akdis M, Akdis CA. Type 2 immunity in allergic diseases. Cell Mol Immunol 2025; 22:211-242. [PMID: 39962262 PMCID: PMC11868591 DOI: 10.1038/s41423-025-01261-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/09/2025] [Indexed: 03/01/2025] Open
Abstract
Significant advancements have been made in understanding the cellular and molecular mechanisms of type 2 immunity in allergic diseases such as asthma, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis (EoE), food and drug allergies, and atopic dermatitis (AD). Type 2 immunity has evolved to protect against parasitic diseases and toxins, plays a role in the expulsion of parasites and larvae from inner tissues to the lumen and outside the body, maintains microbe-rich skin and mucosal epithelial barriers and counterbalances the type 1 immune response and its destructive effects. During the development of a type 2 immune response, an innate immune response initiates starting from epithelial cells and innate lymphoid cells (ILCs), including dendritic cells and macrophages, and translates to adaptive T and B-cell immunity, particularly IgE antibody production. Eosinophils, mast cells and basophils have effects on effector functions. Cytokines from ILC2s and CD4+ helper type 2 (Th2) cells, CD8 + T cells, and NK-T cells, along with myeloid cells, including IL-4, IL-5, IL-9, and IL-13, initiate and sustain allergic inflammation via T cell cells, eosinophils, and ILC2s; promote IgE class switching; and open the epithelial barrier. Epithelial cell activation, alarmin release and barrier dysfunction are key in the development of not only allergic diseases but also many other systemic diseases. Recent biologics targeting the pathways and effector functions of IL4/IL13, IL-5, and IgE have shown promising results for almost all ages, although some patients with severe allergic diseases do not respond to these therapies, highlighting the unmet need for a more detailed and personalized approach.
Collapse
Affiliation(s)
- Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Carina Beha
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Bingjie Zhao
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Can Zeyneloglu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | | | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Asuncion Garcia-Sanchez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Biomedical and Diagnostic Science, School of Medicine, University of Salamanca, Salamanca, Spain
| | - Juan-Felipe Lopez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Li-Li Shi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Minglin Yang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephen Skolnick
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Seed Health Inc., Los Angeles, CA, USA
| | - Raja Dhir
- Seed Health Inc., Los Angeles, CA, USA
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Abhijeet J Kulkarni
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mar Martin-Fontecha
- Departamento de Quimica Organica, Facultad de Optica y Optometria, Complutense University of Madrid, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| |
Collapse
|
42
|
Mannion JM, Rahimi RA. Tissue-Resident Th2 Cells in Type 2 Immunity and Allergic Diseases. Immunol Rev 2025; 330:e70006. [PMID: 39981858 PMCID: PMC11897987 DOI: 10.1111/imr.70006] [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: 08/23/2024] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
Type 2 immunity represents a unique immune module that provides host protection against macro-parasites and noxious agents such as venoms and toxins. In contrast, maladaptive type 2 immune responses cause allergic diseases. While multiple cell types play important roles in type 2 immunity, recent studies in humans and murine models of chronic allergic diseases have shown that a distinct population of tissue-resident, CD4+ T helper type 2 (Th2) cells play a critical role in chronic allergic inflammation. The rules regulating Th2 cell differentiation have remained less well defined than other T cell subsets, but recent studies have shed new light into the specific mechanisms controlling Th2 cell biology in vivo. Here, we review our current understanding of the checkpoints regulating the development and function of tissue-resident Th2 cells with a focus on chronic allergic diseases. We discuss evidence for a barrier tissue checkpoint in initial Th2 cell priming, including the role of neuropeptides, damage-associated molecular patterns, and dendritic cell macro-clusters. Furthermore, we review the evidence for a second barrier tissue checkpoint that instructs the development of multi-cytokine producing, tissue-resident Th2 cells that orchestrate allergic inflammation. Lastly, we discuss potential approaches to therapeutically target tissue-resident Th2 cells in chronic allergic diseases.
Collapse
Affiliation(s)
- Jenny M Mannion
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rod A Rahimi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
43
|
Banaschewski BJH, Michki SN, Sitaraman S, Pan R, Wang JY, Stewart D, Goldy MK, Lin SM, Cantu E, Katzen JB, Basil MC, Emtiazjoo AM, Todd JL, Gokey JJ, Kropski JA, Frank DB, Zepp JA, Young LR. Emergence of inflammatory fibroblasts with aging in Hermansky-Pudlak syndrome associated pulmonary fibrosis. Commun Biol 2025; 8:284. [PMID: 39987372 PMCID: PMC11846979 DOI: 10.1038/s42003-025-07589-9] [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: 08/05/2024] [Accepted: 01/21/2025] [Indexed: 02/24/2025] Open
Abstract
The longitudinal cellular interactions that drive pulmonary fibrosis are not well understood. To investigate the disease underpinnings associated with fibrosis onset and progression, we generated a scRNA-seq atlas of lungs from young and aged mouse models of multiple subtypes of Hermansky-Pudlak syndrome (HPS), a collection of rare autosomal recessive diseases associated with albinism, platelet dysfunction, and pulmonary fibrosis. We have identified an age-dependent increase in SAA3+ inflammatory lung fibroblasts in HPS mice, including in double-mutant HPS1-2 mice which develop spontaneous fibrosis. HPS1 fibroblasts show increased expression of IL-1R1, whereas alveolar type II epithelial cells from HPS2 mice induce the inflammatory gene signature in co-cultured fibroblasts. scRNA-seq of lung tissue from three HPS1 patients similarly shows the presence of inflammatory fibroblasts and increased IL1R1 expression on fibroblasts. These data posit complex interactions between dysfunctional epithelial cells, inflammatory fibroblasts, and recruited immune cells, suggesting potential opportunities for mitigation of the fibrotic cascade.
Collapse
Affiliation(s)
- Brandon J H Banaschewski
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sylvia N Michki
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sneha Sitaraman
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ruby Pan
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joanna Y Wang
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Dominique Stewart
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mary Kate Goldy
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Susan M Lin
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Cantu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeremy B Katzen
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria C Basil
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Amir M Emtiazjoo
- Deparment of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Jamie L Todd
- Duke Clinical Research Institute, Durham, NC, USA
- Duke University Medical Center, Durham, NC, USA
| | - Jason J Gokey
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Veterans Affairs Medical Center, Nashville, TN, USA
| | - David B Frank
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jarod A Zepp
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Lisa R Young
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
44
|
Vieira BM, Paiva MB, Gaspar-Elsas MIC, Xavier-Elsas PP. Coordinated regulation of eosinophil production and migration by glucocorticoids, prostaglandins, and cysteinyl-leukotrienes. Int Immunopharmacol 2025; 148:114067. [PMID: 39823798 DOI: 10.1016/j.intimp.2025.114067] [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: 12/18/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025]
Abstract
INTRODUCTION The spectrum of eosinophil functions has expanded from fighting helminths to multiple novel roles in malignancy, infection, cancer, and metabolism. In asthma, glucocorticoids, prostaglandins (PG), and cysteinyl-leukotrienes (LT) regulate eosinophil biology through separate signaling pathways. Here we've evaluated the complex interplay between Dexa, PGE2, and CysLTs in eosinopoiesis and eosinophil biology in an allergic asthma model. METHODOLOGY We used different inbred mouse strains to probe the interactions between these agents in eosinophil differentiation and maturation in bone marrow culture. Flow cytometry and histological analyses evaluated eosinophil precursor proliferation, maturation, and VLA-4 expression. The in vivo function of eosinophils was assessed by their in vivo migration into allergen-challenged sites. RESULTS Eosinophil production in IL-5-stimulated bone marrow cultures is enhanced by dexamethasone but suppressed by PGE2, which triggers eosinophil apoptosis via inducible NO synthase (iNOS). Dexamethasone-primed cultures contain mostly immature eosinophils; by contrast, dexamethasone associated with PGE2 leads to the production of mature eosinophils (without inducing eosinophil apoptosis), by a mechanism independent of iNOS. Interaction between dexamethasone and LTD4 in culture produces mostly mature eosinophils expressing VLA-4, capable of migration into the lungs in ovalbumin-sensitized and -challenged mice. DISCUSSION The combination of dexamethasone and either PGE2 or LTD4 - both mediators of allergic inflammation - supports the maturation of eosinophils (overcoming the maturation blockade observed with dexamethasone alone), which are functional in an in vivo model of asthma.
Collapse
Affiliation(s)
- Bruno Marques Vieira
- Laboratório de Citocinas Dept. of Immunology Instituto de Microbiologia Prof. Paulo de Góes Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil; Laboratório de Medicina Experimental e Saúde Instituto Oswaldo Cruz FIOCRUZ Rio de Janeiro Brazil; Laboratório de Biomedicina do Cérebro Instituto Estadual do Cérebro Paulo Niemeyer (IECPN) Rio de Janeiro Brazil.
| | - Milla Bezerra Paiva
- Laboratório de Medicina Experimental e Saúde Instituto Oswaldo Cruz FIOCRUZ Rio de Janeiro Brazil
| | | | - Pedro Paulo Xavier-Elsas
- Laboratório de Citocinas Dept. of Immunology Instituto de Microbiologia Prof. Paulo de Góes Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| |
Collapse
|
45
|
Zhang C, Yang T, Yu Y, Jia Q, Xiao WM, Liu S, Yu ZH, Wen CL, Wei Y, Li H, Lü MH. Causal roles of immune cells and metabolites in chronic pancreatitis: a mendelian randomization study. Hereditas 2025; 162:20. [PMID: 39940040 PMCID: PMC11816568 DOI: 10.1186/s41065-025-00378-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/26/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Previous research has established a correlation between immune cells and an increased likelihood of Chronic pancreatitis (CP). However, studies investigating the causal relationship remain limited. METHODS This study utilized publicly available genome-wide association study (GWAS) databases and conducted a two-sample Mendelian randomization (MR) analysis to examine the causal relationships (CRs) among 731 immune cells, 1,400 metabolites, and CP. Mediation MR analysis was also performed to assess whether metabolites serve as mediators in the relationship between immune cells and CP. RESULTS Our study identified four immune cell types that act as risk factors for CP, with odds ratios (OR) ranging between 1.076 and 1.177. In contrast, three immune cell types were found to serve as protective factors, exhibiting OR values between 0.846 and 0.913. Additionally, four metabolites were implicated as risk factors for CP, with OR values ranging from 1.243 to 1.334. On the other hand, eight metabolites were discovered to have a protective effect, with OR values between 0.580 and 0.871. Mediation analysis revealed that cholesterol levels mediate the causal relationship between immune cell cells and CP, with a mediation effect of 0.00918, accounting for 9.18% of the total effect. CONCLUSIONS Our findings provide valuable insights into the genetic underpinnings of CP, highlighting the role of immune cells and plasma metabolites in its pathogenesis. The mediation analysis further suggests that the presence of CD25 on IgD-CD38-B cells may facilitate CP development through the elevation of cholesterol levels. These results not only deepen our understanding of CP but also suggest potential biological targets for therapeutic intervention. Future clinical research should focus on these mediators to develop more effective treatment strategies for CP.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Tao Yang
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yuan Yu
- Gulin County People's Hospital, Luzhou, Sichuan Province, China
| | - Qian Jia
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Wan-Meng Xiao
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Sha Liu
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Ze-Hui Yu
- Laboratory Animal Center, Southwest Medical University, Luzhou, Sichuan, China
- Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Cheng-Li Wen
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yan Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Hao Li
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| | - Mu-Han Lü
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
46
|
Zhai Z, Yang C, Yin W, Liu Y, Li S, Ye Z, Xie M, Song X. Engineered Strategies to Interfere with Macrophage Fate in Myocardial Infarction. ACS Biomater Sci Eng 2025; 11:784-805. [PMID: 39884780 DOI: 10.1021/acsbiomaterials.4c02061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Myocardial infarction (MI), a severe cardiovascular condition, is typically triggered by coronary artery disease, resulting in ischemic damage and the subsequent necrosis of the myocardium. Macrophages, known for their remarkable plasticity, are capable of exhibiting a range of phenotypes and functions as they react to diverse stimuli within their local microenvironment. In recent years, there has been an increasing number of studies on the regulation of macrophage behavior based on tissue engineering strategies, and its regulatory mechanisms deserve further investigation. This review first summarizes the effects of key regulatory factors of engineered biomaterials (including bioactive molecules, conductivity, and some microenvironmental factors) on macrophage behavior, then explores specific methods for inducing macrophage behavior through tissue engineering materials to promote myocardial repair, and summarizes the role of macrophage-host cell crosstalk in regulating inflammation, vascularization, and tissue remodeling. Finally, we propose some future challenges in regulating macrophage-material interactions and tailoring personalized biomaterials to guide macrophage phenotypes.
Collapse
Affiliation(s)
- Zitong Zhai
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Chang Yang
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Wenming Yin
- Department of Neurology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Yali Liu
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong 528000, China
| | - Shimin Li
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Ziyi Ye
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Mingxiang Xie
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Xiaoping Song
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
- Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
47
|
Loperfido A, Cavaliere C, Fionda B, Bellocchi G, Masieri S, Caminati M. Narrative Review of Genetic and Immunological Mechanisms Involved in the Pathogenesis of Kimura's Disease: New Therapeutic Targets. Genes (Basel) 2025; 16:194. [PMID: 40004521 PMCID: PMC11855017 DOI: 10.3390/genes16020194] [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: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Kimura's disease (KD) is a rare, chronic inflammatory disorder that predominantly affects young men of East Asian descent. It is characterized by painless solid masses primarily localized to the deep subcutaneous tissues of the head and neck, eosinophilia, and elevated serum immunoglobulin E (IgE). While the exact cause remains unclear, the pathogenesis is thought to involve dysregulated immune responses, particularly those mediated by T-helper cells 2 (Th2), eosinophils, and IgE production. Advances in molecular biology have suggested that genetic factors play a significant role in the development and progression of this chronic inflammatory condition. Recent studies have implicated several genes and immune pathways in its development, and understanding these genetic components may provide insights into better diagnostic tools and therapeutic strategies for KD. In this regard, biological therapies, by targeting the immune mechanisms underlying KD, have been used to treat this challenging condition with promising results, contributing to a better understanding of the pathogenesis of this rare disorder. The aim of this study was to review the literature concerning the genetic factors and immune mechanisms that contribute to the pathogenesis of KD, with a special focus on the role of biological therapies.
Collapse
Affiliation(s)
- Antonella Loperfido
- Otolaryngology Unit, San Camillo Forlanini Hospital, Circonvallazione Gianicolense 87, 00152 Rome, Italy
| | - Carlo Cavaliere
- Department of Sense Organs, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Bruno Fionda
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Bellocchi
- Otolaryngology Unit, San Camillo Forlanini Hospital, Circonvallazione Gianicolense 87, 00152 Rome, Italy
| | - Simonetta Masieri
- Department of Oral and Maxillofacial Sciences, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Marco Caminati
- Allergy Unit and Asthma Center, Verona Integrated University Hospital, 37134 Verona, Italy
- Department of Medicine, University of Verona, 37124 Verona, Italy
| |
Collapse
|
48
|
Wu X, Pan B, Chu C, Zhang Y, Ma J, Xing Y, Ma Y, Zhu W, Zhong H, Alimu A, Zhou G, Liu S, Chen W, Li X, Puyi S. CXCL16/CXCR6/TGF-β Feedback Loop Between M-MDSCs and Treg Inhibits Anti-Bacterial Immunity During Biofilm Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409537. [PMID: 39716908 PMCID: PMC11831521 DOI: 10.1002/advs.202409537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/29/2024] [Indexed: 12/25/2024]
Abstract
Staphylococcus aureus (S. aureus) is a leading cause of Periprosthetic joint infection (PJI), a severe complication after joint arthroplasty. Immunosuppression is a major factor contributing to the infection chronicity of S. aureus PJI, posing significant treatment challenges. This study investigates the relationship between the immunosuppressive biofilm milieu and S. aureus PJI outcomes in both discovery and validation cohorts. This scRNA-seq analysis of synovium from PJI patients reveals an expansion and heightened activity of monocyte-related myeloid-derived suppressor cells (M-MDSCs) and regulatory T cells (Treg). Importantly, CXCL16 is significantly upregulated in M-MDSCs, with its corresponding CXCR6 receptor also elevated on Treg. M-MDSCs recruit Treg and enhance its activity via CXCL16-CXCR6 interactions, while Treg secretes TGF-β, inducing M-MDSCs proliferation and immunosuppressive activity. Interfering with this cross-talk in vivo using Treg-specific CXCR6 knockout PJI mouse model reduces M-MDSCs/Treg-mediated immunosuppression and alleviates bacterial burden. Immunohistochemistry and recurrence analysis show that PJI patients with CXCR6high synovium have poor prognosis. This findings highlight the critical role of CXCR6 in Treg in orchestrating an immunosuppressive microenvironment and biofilm persistence during PJI, offering potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Baiqi Pan
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Chenghan Chu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Yangchun Zhang
- Department of OrthopedicsThe People's Hospital of Baoan ShenzhenShenzhenGuangdong518101China
- Department of OrthopedicsThe Second Affiliated Hospital of Shenzhen UniversityShenzhenGuangdong518101China
| | - Jinjin Ma
- Technology School of MedicineSouth China University of TechnologyGuangzhouGuangdong510640China
- Shien‐ming Wu School of Intelligent EngineeringSouth China University of TechnologyGuangzhouGuangdong510640China
| | - Yang Xing
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Yuanchen Ma
- Department of OrthopedicsGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdong519041China
| | - Wengang Zhu
- Department of Joint OrthopedicsYuebei People's HospitalShaoguanGuangdong512099China
| | - Huan Zhong
- Department of Joint SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong524002China
| | - Aerman Alimu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Guanming Zhou
- Department of OrthopedicsFoshan Hospital of Traditional Chinese MedicineGuangzhouGuangdong528051China
| | - Shuying Liu
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Weishen Chen
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Xiang Li
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of Spine SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Sheng Puyi
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| |
Collapse
|
49
|
Liu MK, Tang JJ, Li H, Chen XY, Cai JL, Lin GY, Chen KY, Liu ZP, Ji XF, Yang ZJ, Li Z. Artemisitene ameliorates carbon tetrachloride-induced liver fibrosis by inhibiting NLRP3 inflammasome activation and modulating immune responses. Int Immunopharmacol 2025; 146:113818. [PMID: 39681062 DOI: 10.1016/j.intimp.2024.113818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024]
Abstract
Artemisitene (ATT), an artemisinin (ART) analog retaining the endoperoxide moiety and incorporating an additional α, β-unsaturated carbonyl structure, exhibits enhanced biological activities. However, its therapeutic effects on liver fibrosis remain unclear. In this study, we demonstrated that ATT significantly alleviated liver inflammation and fibrosis induced by carbon tetrachloride (CCL4) in mice. ATT treatment markedly reduced the count of neutrophils in the liver, as well as macrophages in both the liver and spleen. Additionally, the frequencies of Th2 and Th17 cells were significantly lowered, while Th1 cells frequency and the Th1/Th2 index were notably increased. The frequency of ILC2 cells, correlated with ST2 and IL-33 expression levels, was also significantly lowered. Consistently, ATT inhibited NLRP3 inflammasome activation, which was positively associated with AST and ALT levels, and with the count of Neutrophils, macrophages, and ILC2 cells, but negatively correlated with Th1frequeny. Furthermore, liver fibrosis severity showed a significant positive correlation with neutrophil and Th17 cell counts in the liver, and a negative correlation with Th1 cell count and the Th1/Th2 index. Therefore, ATT alleviated CCL4-induced mice liver fibrosis through NLRP3 inflammasome inhibition and immunomodulation.
Collapse
Affiliation(s)
- Meng-Ke Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Juan-Juan Tang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Hao Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xu-Yang Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jun-Ling Cai
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Gui-Ying Lin
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Kan-Yao Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; Department of Clinical Laboratory, Guangdong Provincial People's Hospital Zhuhai Hospital (Zhuhai Golden Bay Center Hospital), Zhuhai, China
| | - Zhi-Peng Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Affiliated Qingyuan Hospital of Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, China
| | - Xiao-Fang Ji
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhong-Jin Yang
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, China.
| | - Zi Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
50
|
Igarashi Y, Wada H, Muto M, Sone R, Hasegawa Y, Seino KI. Amelioration of liver fibrosis with autologous macrophages induced by IL-34-based condition. Inflamm Regen 2025; 45:2. [PMID: 39856797 PMCID: PMC11758727 DOI: 10.1186/s41232-025-00364-7] [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: 08/26/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND For the treatment of liver fibrosis, several novel cell therapies have been proposed. Autologous macrophage therapy has been reported as one of the promising treatments. So far, most studies have used colony-stimulating factor 1 (CSF-1) to induce the differentiation of macrophage progenitor cells. The receptor for CSF-1, CSF-1R possesses another ligand, interleukin 34. However, the therapeutic capacity for liver fibrosis by interleukin 34-induced macrophages has not been evaluated. METHODS We have employed acute (bile duct ligation) and chronic (administration of carbon tetrachloride or thioacetamide) liver fibrosis models. Using these models, we evaluated the therapeutic capacity of macrophages induced by interleukin 34-based conditions. In most experiments, interleukin 4 was also added to the differentiation process to induce alternative-activated macrophages. As a mechanism analysis, we have examined liver inflammation and damage, the status of stellate cells, and the immunosuppressive capacity of the macrophages. Human macrophages were differentiated from CD14+ monocytes and analyzed. RESULTS In both acute and chronic liver damage experiments, interleukin 34-induced macrophages significantly ameliorated liver fibrosis. The addition of interleukin 4 to the differentiation process resulted in an increase of obtained macrophages and a bias to alternative activated macrophages (so-called M2). The alternative activated macrophages (M2-type) showed a reproducible therapeutic effect of liver fibrosis with a suppression of parameters of liver inflammation and damage, stellate cells, and T cell activation. Similar macrophages could be differentiated from human CD14+ monocytes in the presence of interleukin 34 plus interleukin 4, and a therapeutic effect was observed using a humanized mouse model. CONCLUSIONS Interleukin 34-induced macrophages, particularly when additionally stimulated with interleukin 4, significantly ameliorated the liver fibrosis.
Collapse
Affiliation(s)
- Yuichi Igarashi
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Haruka Wada
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Masato Muto
- MEDINET Medical Institute, MEDINET Co., Ltd., Tokyo, Japan
| | - Ryohei Sone
- MEDINET Medical Institute, MEDINET Co., Ltd., Tokyo, Japan
| | - Yoshinori Hasegawa
- Laboratory of Gene Sequencing Analysis, Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Ken-Ichiro Seino
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|