1
|
Lyu Q, Kouketsu Y, Tazaki A, Kato M, Motooka Y, Toyokuni S. Terrestrial iron sulfide minerals induce distinct regulation of intracellular redox homeostasis and iron assimilation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118327. [PMID: 40381394 DOI: 10.1016/j.ecoenv.2025.118327] [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: 11/12/2024] [Revised: 03/12/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Repeated exposure to airborne terrestrial natural minerals may cause pneumoconiosis and lung cancer, among which iron sulfide is identified as an aggravating factor. In the biological system, iron-sulfur cluster is an inorganic cofactor that is evolutionarily conserved in all the living organisms. Whereas ferrous iron catalyzes the generation of hydroxyl radicals, sulfur is indispensable as a component of antioxidants, such as glutathione. Imbalanced redox homeostasis contributes to oxidative stress, causing ferroptosis, an iron-dependent regulated necrosis characterized by lipid peroxidation, resulting in various disorders. We undertook this study to understand the cellular regulatory mechanisms against major terrestrial minerals containing iron and sulfur from the viewpoint of cellular redox. We used fundamental iron sulfide minerals collected from natural sources to treat human macrophage and fibroblast cells and investigated the biological responses. Alterations in sulfane sulfur, glutathione and iron have been analyzed using either specific fluorescent probes or inductively coupled plasma mass spectrometry. Iron sulfide microparticles with high Fe/S ratio (pyrrhotite; Fe1-XS) induced more reactive sulfane species and glutathione, with less catalytic iron inside cells, whereas the mineral with low Fe/S ratio (pyrite; FeS2) exhibited the opposite effects. Notably both showed cytotoxicity, where pyrite caused ferroptosis but pyrrhotite led to non-ferroptotic disruption. Furthermore, assimilated cellular excess iron was secreted via CD63(+) exosome containing iron-loaded ferritin to the extracellular space with higher iron content in pyrrhotite. Our findings suggest that iron and sulfur work complementarily in maintaining intracellular redox homeostasis, which would be crucial to understand the associated pathology.
Collapse
Affiliation(s)
- Qinying Lyu
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yui Kouketsu
- Department of Earth and Planetary Sciences, Nagoya University Graduate School of Environmental Studies, Furo-cho, Chikusa, Nagoya 484-8601, Japan
| | - Akira Tazaki
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Activity of the Institute of Innovation for Future Society of Nagoya University, Japan
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Activity of the Institute of Innovation for Future Society of Nagoya University, Japan
| | - Yashiro Motooka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Center for Low Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8603, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Furo-Cho, Chikusa-ku, Nagoya 464-8603, Japan.
| |
Collapse
|
2
|
Little I, Bersie S, Redente EF, McCubbrey AL, Tarling EJ. Alveolar macrophages: guardians of the alveolar lipid galaxy. Curr Opin Lipidol 2025; 36:153-162. [PMID: 40183504 PMCID: PMC12043416 DOI: 10.1097/mol.0000000000000987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
PURPOSE OF REVIEW As the primary guardians at the air-surface interface, the functional profile of alveolar macrophages (AM) is wide-ranging from establishment of the alveolar niche, homeostatic maintenance of surfactant levels, to pathogen clearance and resolution and repair processes. Alveolar lipid homeostasis is disturbed in chronic lung diseases and contributes to disease pathogenesis through extracellular localization in the alveolar lumen or intracellular accumulation in AM. This review aims to provide a focused overview of the state of knowledge of AM, their ontogeny and development during health and disease, and how dysregulated AM lipids play a key role in disease processes, from initiation to resolution. RECENT FINDINGS While lipid-laden macrophages are observed across a broad spectrum of lung diseases, their occurrence has largely been considered consequential. Recent advances in lipidomic profiling of single cell types has revealed that disturbances to lipid homeostasis occur early in disease in tissue-resident cells. Comparisons between inflammatory and fibrotic injury models reveal specific alveolar macrophage subsets with different lipid utilization that contribute to the disease process. SUMMARY Understanding the intricate web of AM population seeding and development and how this niche is perturbed by lipid disturbances may help provide leverage for new interventions.
Collapse
Affiliation(s)
- Isaiah Little
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Stephanie Bersie
- Toxicology Graduate Program, Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Elizabeth F. Redente
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pediatrics
| | - Alexandra L. McCubbrey
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Elizabeth J. Tarling
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Molecular Biology Institute
- Johnsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
3
|
Yang S, Penna V, Lavine KJ. Functional diversity of cardiac macrophages in health and disease. Nat Rev Cardiol 2025; 22:431-442. [PMID: 39743564 DOI: 10.1038/s41569-024-01109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Macrophages make up a substantial portion of the stromal compartment of the heart in health and disease. In the past decade, the origins of these cardiac macrophages have been established as two broad populations derived from either embryonic or definitive haematopoiesis and that can be distinguished by the expression of CC-motif chemokine receptor 2 (CCR2). These cardiac macrophage populations are transcriptionally distinct and have differing cell surface markers and divergent roles in cardiac homeostasis and disease. Embryonic-derived CCR2- macrophages are a tissue-resident population that participates in tissue development, repair and maintenance, whereas CCR2+ macrophages are derived from definitive haematopoiesis and contribute to inflammation and tissue damage. Studies from the past 5 years have leveraged single-cell RNA sequencing technologies to expand our understanding of cardiac macrophage diversity, particularly of the monocyte-derived macrophage populations that reside in the injured and diseased heart. Emerging technologies in spatial transcriptomics have enabled the identification of distinct disease-associated cellular neighbourhoods consisting of macrophages, other immune cells and fibroblasts, highlighting the involvement of macrophages in cell-cell communication. Together, these discoveries lend new insights into the role of specific macrophage populations in the pathogenesis of cardiac disease, which can pave the way for the identification of new therapeutic targets and the development of diagnostic tools. In this Review, we discuss the developmental origin of cardiac macrophages and describe newly identified cell states and associated cellular neighbourhoods in the steady state and injury settings. We also discuss various contributions and effector functions of cardiac macrophages in homeostasis and disease.
Collapse
Affiliation(s)
- Steven Yang
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Vinay Penna
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
4
|
Li G, He W, Wang DW. Immune cell dynamics in heart failure: implicated mechanisms and therapeutic targets. ESC Heart Fail 2025; 12:1739-1758. [PMID: 39905753 PMCID: PMC12055366 DOI: 10.1002/ehf2.15238] [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: 10/24/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
The relationship between heart failure (HF) and immune activation has garnered significant interest. Studies highlight the critical role of inflammation in HF, affecting cardiac structure and function. Despite promising anti-inflammatory therapies, clinical trials have faced challenges, indicating an incomplete understanding of immune mechanisms in HF. Immune cells, which are key cytokine sources, are pivotal in HF progression. In this review, the authors provide a comprehensive overview of the complex role of different types of immune cells and their cell subtypes in HF. In addition, the authors summarize the available targets and animal experimental evidence for targeting immune cells for the treatment of HF. Future research directions will focus on the roles of immune cells and their interrelationships at different stages of HF, aiming to develop more targeted therapeutic strategies that can achieve more precise interventions in the pathological process of HF.
Collapse
Affiliation(s)
- Gen Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| | - Wu He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| |
Collapse
|
5
|
Habash SA, Takahashi N, Eltalkhawy YM, Abdelnaser RA, Ogata-Aoki H, Okada S, Takizawa H, Usuki S, Etoh K, Hino S, Morino-Koga S, Ogawa M, Suzu S. Macrophages with different origins proliferate ex vivo and do not lose their core intrinsic features. iScience 2025; 28:112635. [DOI: 10.1016/j.isci.2025.112635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2025] Open
|
6
|
Zhang SP, Tong M, Mo J, Dong ZY, Huang YF. M2 macrophages activate the IL-10/JAK2/STAT3 pathway to induce pathological microangiogenesis in the nucleus pulposus exacerbating intervertebral disc degeneration. J Orthop Surg Res 2025; 20:532. [PMID: 40426248 PMCID: PMC12117970 DOI: 10.1186/s13018-025-05962-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Accepted: 05/23/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Macrophage infiltration accompanied by pathological microangiogenesis in the nucleus pulposus (NP) plays a critical role in the progression of intervertebral disc degeneration (IDD). However, the involvement of M2 macrophages in mediating NP pathological angiogenesis and their underlying mechanisms remain unclear. METHODS Firstly, the expression of M2 macrophage (CD206) and microangiogenic (CD34) markers in human degenerated NP was observed by immunohistochemical staining, subsequently, a co-culture system of M2 macrophages and NP cells was established. IL-10 expression was silenced using siRNA to assess the pro-angiogenic effects of M2 macrophages in IDD via IL-10 and its downstream janus kinase (JAK) 2/ signal transducer and activator of transcription (STAT) 3 pathway. AG490, a specific JAK2/STAT3 inhibitor, was applied to determine whether IL-10 exerts its effects through this pathway and to evaluate its impact on angiogenesis and extracellular matrix (ECM) metabolism in NP pathology. RESULTS CD206 and CD34 were co-expressed in degenerated NP tissue. Degenerated NP cells secreted CCL17, CCL18, and CD206, exhibiting M2-like characteristics. Co-culture of M2 macrophages with degenerated NP cells led to IL-10 secretion to promote CD34 expression, and downregulated anabolic genes (type II collagen (COL2), aggrecan), and upregulated catabolic genes (matrix metalloproteinase (MMP)-3, MMP-7). JAK2 and STAT3 expression was significantly increased following co-culture. Activation of the JAK2/STAT3 pathway enhanced vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor (VEGFR), and CD34 expression and induced further downregulation of COL2 and aggrecan and upregulation of MMP-3 and MMP-7. CONCLUSION M2 macrophage infiltration and pathological neovascularization are prominent in degenerated NP tissue. IL-10 secreted by M2 macrophages activates the JAK2/STAT3 pathway to promote pathological microangiogenesis by up-regulate the expression of VEGF/VEGFR. This process disrupts ECM and accelerates the progression of IDD. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Si-Ping Zhang
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China
| | - Min Tong
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China
| | - Jun Mo
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China
| | - Zhen-Yu Dong
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China
| | - Yi-Fei Huang
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China.
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China.
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China.
| |
Collapse
|
7
|
Bigenwald C, Roos-Weil D, Pagès A, Hélias-Rodzewicz Z, Copie-Bergman C, Nashvi M, Khneisser P, Parrens M, Traverse-Glehen A, Ray-Coquard I, Ysebaert L, Marchand T, Razanamahery J, Charlotte F, Neel A, Damaj G, Dion J, Nazal-Traissac EM, Tardy S, Salmeron G, Monjanel H, Idbaih A, Héritier S, Haroche J, Donadieu J, Emile JF. Characterization and treatment outcomes of malignant histiocytoses in a retrospective series of 141 cases in France. Blood Adv 2025; 9:2530-2541. [PMID: 40009752 DOI: 10.1182/bloodadvances.2024015208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
ABSTRACT Malignant histiocytoses (MH) are rare and poorly understood cancers, with no established therapeutic guidelines. We conducted a national retrospective study of MH diagnosed in France between 2000 and 2023. All cases underwent centralized histological review, and several malignant tumors with a stroma highly enriched in histiocytes were excluded. In total, 141 patients were included, with a median age of 62 years (range, 1-87). The cases comprised either primary MH (64%) or MH associated with other hematologic malignancies (36%). Phenotypes corresponded to histiocytic (43%), interdigitating dendritic cell (37%) or Langerhans cell (12%) sarcomas, or high-grade indeterminate dendritic cell tumors (10%), as per the World Health Organization classification. Tumor cells were almost universally positive for CSF1R and PU.1, and 85% showed phosphorylated extracellular signal-regulated kinase positivity. Next-generation sequencing was performed in 75 cases. Mutations in the MAPK pathway were more frequent in secondary compared with primary MH (90% vs 55%; P = .0012). PTPN11 mutations were exclusively observed in primary MH (P = .0035). Mutations in genes related to DNA methylation mechanisms (TET2, ASXL1, DNMT3A) and TP53 were present in 20% and 14% of cases, respectively. Although therapeutic regimens varied considerably, our results demonstrate that surgical resection in localized cases, and the use of BRAF or MEK inhibitors achieved the highest complete response rates, at 63% and 21%, respectively. The prognosis remains poor, with a 5-year overall survival rate of 31%, which is comparable to that of T/natural killer cell lymphomas. Prospective follow-up and a standardized treatment approach in specialized reference centers are crucial to improving patient survival. This trial was registered at www.clinicaltrials.gov as #NCT04437381.
Collapse
Affiliation(s)
- Camille Bigenwald
- Département d'Hématologie, Gustave Roussy, Villejuif, France
- INSERM U1015, Gustave Roussy, Villejuif, France
| | - Damien Roos-Weil
- Sorbonne Université, Hematology Unit, Centre Hospitalier Universitaire Pitié-Salpêtrière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Arnaud Pagès
- Bureau de Biostatistique et d'Epidémiologie, Gustave Roussy et Centre de Recherche en Epidémiologie et Santé des Populations U1018, Oncostat, labellisé Ligue Contre le Cancer, INSERM, Villejuif, France
| | - Zofia Hélias-Rodzewicz
- Paris-Saclay University, Versailles Saint Quentin en Yvelynes University, EA4340-Biomarqueurs et Essais Cliniques en Cancérologie et Onco-Hématologie, Assistance Publique Hôpitaux de Paris, Ambroise-Paré Hospital, Smart Imaging, Service de Pathologie, Boulogne, France
| | - Christiane Copie-Bergman
- Paris-Est Créteil University, INSERM, Institut Mondor de Recherche Biomédicale, Pathology Department, Assistance Publique des Hôpitaux de Paris, Centre Hospitalier Universitaire Henri Mondor, Créteil, France
| | | | - Pierre Khneisser
- Department of Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Marie Parrens
- Pathology Department, University of Bordeaux, Haut-Lévêque Hospital, Pessac, France
| | - Alexandra Traverse-Glehen
- Unité de Formation et de Recherche Claude Bernard Lyon 1, Hospices Civils de Lyon, Institut de Pathologie Multisite, Hôpital Lyon Sud, Lyon, France
| | - Isabelle Ray-Coquard
- Université Claude Bernard Lyon 1, Hesper Laboratory, Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Loïc Ysebaert
- Hematology Department, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Tony Marchand
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire de Rennes, Universitéde Rennes, Unité Mixte de Recherche 1236, INSERM, Établissement Français du Sang Bretagne, Rennes, France
| | - Jérôme Razanamahery
- Department of internal medicine and clinical immunology, Dijon University Hospital, Dijon, France
| | - Frédéric Charlotte
- Pathology Department, Pitié Salpêtrière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Antoine Neel
- Service de Médecine Interne, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Gandhi Damaj
- Institut d'Hématologie, Centre Hospitalier Universitaire de Caen, Université de Caen-Normandie, Caen, France
| | - Jérémie Dion
- Internal Medicine and Immunopathology Department, University Hospital Center of Toulouse Oncopole, Toulouse, France
| | | | - Stéphanie Tardy
- Departement d'Hématologie, Centre Hospitalier Annecy Genevois, Annecy, France
| | - Géraldine Salmeron
- Département d'Hématologie, Centre Hospitalier Intercommunal de Poissy, Saint Germain en Laye, France
| | - Hélène Monjanel
- Service de Médecine Interne, Centre Hospitalier Henri Mondor d'Aurillac, Aurillac, France
| | - Ahmed Idbaih
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique, Unité Mixte de Recherche S 1127, Institut du Cerveau et de la Moelle Épinière, Institut du Cerveau et de la Moelle, Assistance Publique Hôpitaux de Paris, Hôpitaux Universitaires La Pitié Salpêtrière Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - Sébastien Héritier
- French Reference Center for Histiocytosis, Department of Pediatric Hematology and Oncology, Trousseau Hospital, Assistance Publique Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Julien Haroche
- Sorbonne University, Internal Medicine Department 2, Institut E3M, French Reference Centre for Histiocytosis, Pitié-Salpȇtrière Hospital, Centre d'Immunologie et des Maladie Infectieuses INSERM-UMRS 1135, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jean Donadieu
- French Reference Center for Histiocytosis, Department of Pediatric Hematology and Oncology, Trousseau Hospital, Assistance Publique Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Jean-François Emile
- Paris-Saclay University, Versailles Saint Quentin en Yvelynes University, EA4340-Biomarqueurs et Essais Cliniques en Cancérologie et Onco-Hématologie, Assistance Publique Hôpitaux de Paris, Ambroise-Paré Hospital, Smart Imaging, Service de Pathologie, Boulogne, France
| |
Collapse
|
8
|
Feng Y, Kong J, Sun W, Li Y, Ren F, Sun X, Li M, Liu Y, Sun S, Qin H. NLRP3 regulates macrophage function by M-CSF/M-CSFR signaling in acute radiation-induced lung injury. Mol Immunol 2025; 183:213-224. [PMID: 40403643 DOI: 10.1016/j.molimm.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/25/2025] [Accepted: 05/11/2025] [Indexed: 05/24/2025]
Abstract
Alveolar macrophages are the most abundant macrophages in the healthy lungs and are important players in maintaining lung homeostasis as well as orchestrating tissue repair after injury. Many studies have proved that the initiation, development and progression of acute radiation-induced lung injury are associated with alveolar macrophages. However, lung-associated macrophages function and developmental processes in acute radiation-reduced lung injury remain elusive. To investigate the role of NLRP3 in radiation-reduced lung injury, we established wild-type and NLRP3-/- mice models, and we found that the extent of pneumonia reduced in NLRP3-/- IR group. In in vivo experiments, we observed a decrease in the number of macrophages in NLRP3-/- group. At the same time, in in vitro experiments we have found that macrophages are more easily polarized toward the M2 after radiation in NLRP3-/- group compared with the control group. Our findings reveal that NLRP3 affects the differentiation and chemotaxis of alveolar macrophages through M-CSF/M-CSFR signalling at the onset of radiation-induced lung injury.
Collapse
Affiliation(s)
- Yuqing Feng
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Jiao Kong
- China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Wanyue Sun
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Yan Li
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Fule Ren
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Xuetong Sun
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Mingxi Li
- China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Ying Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Shilong Sun
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Haiyan Qin
- China-Japan Union Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
9
|
Liu Y, Wang TT, Lu Y, Riaz M, Qyang Y. Cardiac macrophage: Insights from murine models to translational potential for human studies. J Mol Cell Cardiol 2025; 204:17-31. [PMID: 40354877 DOI: 10.1016/j.yjmcc.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Macrophages are a cell type that are known to play dynamic roles in acute and progressive pathology. They are highly attuned to their microenvironments throughout maturation, tailoring their functional responses according to the specific tissues in which they reside and their developmental origin. Cardiac macrophages (cMacs) have emerged as focal points of interest for their interactions with the unique electrical and mechanical stimuli of the heart, as well as for their role in maintaining cardiac homeostasis. Through an in-depth analysis of their origin, lineage, and functional significance, this review aims to shed light on cMacs' distinct contributions to both normal physiological maintenance as well as disease progression. Central to our discussion is the comparison of cMac characteristics between mouse and human models, highlighting current challenges and proposing novel experimental tools for deciphering cMac function within the intricate human cardiac microenvironments based on current murine studies. Our review offers valuable insights for identifying novel therapeutic targets and interventions tailored to the distinct roles of these immune cells in cardiovascular diseases (CVDs).
Collapse
Affiliation(s)
- Yufeng Liu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Yale Biological and Biomedical Sciences, Graduate School of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Tricia T Wang
- Yale Biological and Biomedical Sciences, Graduate School of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Yinsheng Lu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Yale Stem Cell Center, New Haven, CT, USA; Department of Mechanical Engineering and Materials Science, Graduate School of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Muhammad Riaz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
10
|
Ho MFS, Farkas O, Faria AV, Plemel JR, Kerr BJ. A recent history of immune cell sex differences in the peripheral nervous system in persistent pain states. Brain Behav Immun 2025; 128:766-775. [PMID: 40345628 DOI: 10.1016/j.bbi.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025] Open
Abstract
Pain is entwined with inflammation, and biological sex often influences mechanisms of the immune system. Due to possible differences in inflammatory mechanisms, women are predisposed to autoimmune diseases and chronic pain. Despite sex as a critical variable in clinical cases of autoimmune conditions and its pain comorbidities, fundamental investigations have long underrepresented female subjects in their studies. Fundamental research in the 2010s, however, identified a binary sex specific mechanism for pain in rodents: male pain is microglia-driven while female pain is T cell-driven. Since then, studies have expanded in neuro-immunology to indicate that the sex differences and immune cells involved in these processes take on more elaborate roles when expanded to other causal modalities and anatomical levels of neuropathic and inflammatory pain. In this mini-review, we highlight updated roles for macrophages, T cells, and B cells in the peripheral nervous system during persistent pain conditions: neuropathic pain and inflammatory pain. We discuss sex similarities and sex differences in these cell types. By parsing out the sex specific roles of immune cells in persistent pain states, we may be better positioned to find immune-based therapies that can effectively target chronic pain in sex-biased autoimmune conditions.
Collapse
Affiliation(s)
- Madelene Faye S Ho
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | - Olivia Farkas
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Andre Vilela Faria
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Pharmacology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Physiology, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
11
|
Joldes C, Jimbu L, Mesaros O, Zdrenghea M, Fetica B. Tumor-Associated Macrophages as Key Modulators of Disease Progression in Diffuse Large B-Cell Lymphoma. Biomedicines 2025; 13:1099. [PMID: 40426926 PMCID: PMC12108958 DOI: 10.3390/biomedicines13051099] [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: 02/09/2025] [Revised: 04/17/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025] Open
Abstract
With the advent of new therapeutic approaches, there is hope that anticancer treatment will eventually be possible without the use of chemotherapy. Efficient immunotherapeutic options have recently emerged in many cancers, offering a less aggressive approach, with overall better tolerance, making them also suitable for frail patients. Response to immunotherapy relies on the availability, functionality, and efficacy of the host's immune effector mechanisms. One of the key factors determining the efficacy of immunotherapy is the tumor microenvironment, which encompasses various immune effectors, including macrophages, which play a crucial role in regulating immune responses through phagocytosis and antigen presentation. Macrophages are prototypically divided, according to their polarization, into either the pro-inflammatory M1 type or the anti-inflammatory M2 type. In the tumor microenvironment, M2-polarized macrophages, known as tumor-associated macrophages (TAMs), are the predominant phenotype and are associated with tumor progression. The M1/M2 paradigm contributes to the understanding of tumor progression. Due to the variable microenvironment, the mechanisms regulating TAMs can vary across different cancers. Variations in TAM polarization may account for the different treatment responses in patients with similar diseases. This paper investigates the connection between TAMs, disease progression, and treatment responses in the most frequent solid hematologic cancer, diffuse large B-cell lymphoma.
Collapse
Affiliation(s)
- Corina Joldes
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
| | - Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Bogdan Fetica
- Department of Pathology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania;
| |
Collapse
|
12
|
Grabowski GA, Kishnani PS, Alcalay RN, Prakalapakorn SG, Rosenbloom BE, Tuason DA, Weinreb NJ. Challenges in Gaucher disease: Perspectives from an expert panel. Mol Genet Metab 2025; 145:109074. [PMID: 40112481 DOI: 10.1016/j.ymgme.2025.109074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/25/2024] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
This focused review concentrates on eight topics of high importance for Gaucher disease (GD) clinicians and researchers: 1) The consideration of GD as distinct types rather than a spectrum. A review of the literature clearly supports the view that there are distinct types of GD. Type 1 is characterized by the absence of primary neuronopathic involvement, while types 2 and 3 are characterized by progressive primary neuronopathic disease. 2) Neurologic and neuronopathic manifestations. A growing body of evidence indicates that the peripheral nervous system may be involved in GD type 1 and that there may also be signs and symptoms of central nervous system (CNS) disease in this group. However, GD type 1 is characterized by the absence of primary neuronopathic disease, whereas GD types 2 and 3 are characterized by progressive, albeit variable, primary neuronopathic disease. Abnormalities in saccadic eye movements have been suggested as being diagnostic for neuronopathic GD, but they may also occur in GD type 1 and in other inflammatory diseases. 3) The importance of whole GBA1 sequencing. This approach is superior to exome sequencing because of potential effects of deep intronic variants on gene expression. It also has the capacity to detect variant alleles that might be missed with gene panels. 4) Monoclonal gammopathy of undetermined significance (MGUS). The risks of MGUS, multiple myeloma, and non-Hodgkin's lymphoma are elevated in patients with GD compared to the general population and strong evidence indicates that lyso-Gb1 stimulates the formation of monoclonal immunoglobulins (M-protein) in patients with GD and MGUS. 5) Pulmonary involvement in GD. Pulmonary complications can be identified through spirometry in up to 45 % of patients with GD type 1 and 55 % of those with GD type 3. Limited evidence exists that enzyme replacement therapy (ERT) reduces the severity of these complications in patients with GD type 1. 6) Gaucheromas. These may occur in patients with GD types 1 or 3, but there is little detailed information about their inception, mechanisms underlying growth, cellular organization, and biochemical activities, and no definitive guidance for their management. Gaucheromas behave like benign (i.e. non-metastasizing) neoplasms, and it may be reasonable to classify them as such. 7) Bone and joint involvement. Dual-energy X-ray absorptiometry scans alone are insufficient for monitoring all changes in bone that may occur in patients with GD. Quantitative magnetic resonance imaging (MRI) techniques using Dixon quantitative chemical shift imaging have provided results that correlate with GD severity scores, bone complications, and biomarkers for GD bone involvement. Thoracic kyphosis is a common complication of GD types 1 and 3, and there is very limited information regarding the effects of ERT or substrate synthesis inhibition therapy (SSIT) on this condition. 8) Treatment initiation, selection, combination, and switching. Prompt initiation of treatment in pediatric patients is important as GD can lead to impaired growth, lower peak bone mass, and delayed puberty. These adverse outcomes can often be ameliorated or prevented with timely treatment. Either ERT or eliglustat, a SSIT agent, is suitable as first-line treatment of adults with GD. Studies of switching from ERT to eliglustat, or between different ERT products, have indicated that changing treatment is safe, although efficacy outcomes vary. A critical remaining issue is the lack of treatments capable of reaching the CNS to slow or halt the progression of neuronopathic disease in patients with GD type 2 or 3 and potentially reduce the risk of Parkinson's disease in GD type 1 patients and heterozygotes for GBA1 variants.
Collapse
Affiliation(s)
- Gregory A Grabowski
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH 45267, USA; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, 905 Lasalle Street, GSRB1, 4th Floor, Room 4010, Durham, NC 27710, USA.
| | - Roy N Alcalay
- Neurological Institute of New York, Columbia University, 710 West 168th Street, New York, NY 10032, USA.
| | - S Grace Prakalapakorn
- Department of Ophthalmology and Pediatrics, Duke University Medical Center, 2351 Erwin Rd, Box 3802, DUMC, Durham, NC 27705-4699, USA.
| | - Barry E Rosenbloom
- Cedars-Sinai Tower Hematology Oncology Medical Group, 9090 Wilshire Blvd #300, Beverly Hills, CA 90211, USA.
| | - Dominick A Tuason
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, 800 Howard Ave, New Haven, CT 06510, USA.
| | - Neal J Weinreb
- University of Miami UHealth Sylvester Cancer Center Coral Springs, 8170 Royal Palm Blvd, Coral Springs, FL 33065, USA
| |
Collapse
|
13
|
Fei X, Li N, Xu X, Zhu Y. Macrophage biology in the pathogenesis of Helicobacter pylori infection. Crit Rev Microbiol 2025; 51:399-416. [PMID: 39086061 DOI: 10.1080/1040841x.2024.2366944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
Infection with H. pylori induces chronic gastric inflammation, progressing to peptic ulcer and stomach adenocarcinoma. Macrophages function as innate immune cells and play a vital role in host immune defense against bacterial infection. However, the distinctive mechanism by which H. pylori evades phagocytosis allows it to colonize the stomach and further aggravate gastric preneoplastic pathology. H. pylori exacerbates gastric inflammation by promoting oxidative stress, resisting macrophage phagocytosis, and inducing M1 macrophage polarization. M2 macrophages facilitate the proliferation, invasion, and migration of gastric cancer cells. Various molecular mechanisms governing macrophage function in the pathogenesis of H. pylori infection have been identified. In this review, we summarize recent findings of macrophage interactions with H. pylori infection, with an emphasis on the regulatory mechanisms that determine the clinical outcome of bacterial infection.
Collapse
Affiliation(s)
- Xiao Fei
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nianshuang Li
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xinbo Xu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
14
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
15
|
Chen YF, Lu YQ, Gao WY, Fan BY, Ren FC, Shen CP. Anti-inflammatory abietane-type diterpenoids from the roots of Salvia przewalskii. PHYTOCHEMISTRY 2025; 237:114523. [PMID: 40311889 DOI: 10.1016/j.phytochem.2025.114523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/03/2025]
Abstract
Fourteen diterpenes were isolated and purified from the roots of Salvia przewalskii, including eight previously unreported abietane analogues (compounds 1-4, 9, 10a, 11a, and 11b), five known ones (5-8 and 10b), and one icetexane analogue (12). The structures were determined through spectroscopic data interpretation, optical rotations, calculated NMR-DP4+ analysis, and ECD. Compounds 1-8 belong to a class of abietane derivatives containing a [5, 5]-oxospirolactone moiety. The research explored the mechanism behind the prevalence of 16β-type [5, 5]-oxospirolactone as the major component in the inseparable mixtures, shedding light on the proposed biosynthetic pathway of these compounds. Consistent with experimental findings, it was revealed that the 16β-type [5, 5]-oxospirolactone was shown to exhibit significantly lower free energy of formation compared to the 16α-product. Additionally, all compounds were evaluated for their ability to inhibit NO production in LPS-induced RAW 264.7 macrophages. Compounds 1, 2, and 11a demonstrated promising bioactive properties in terms of inhibiting NO release.
Collapse
Affiliation(s)
- Yan-Fang Chen
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Yu-Qing Lu
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Wan-Yun Gao
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Bo-Yi Fan
- School of Pharmacy, Nantong University, 9 Seyuan Road, Nantong, 226019, People's Republic of China
| | - Fu-Cai Ren
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China.
| | - Chuan-Pu Shen
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China.
| |
Collapse
|
16
|
De T, Coin L, Herberg J, Johnson MR, Järvelin MR. Plasma metabolomic signatures for copy number variants and COVID-19 risk loci in Northern Finland populations. Sci Rep 2025; 15:13172. [PMID: 40240424 PMCID: PMC12003712 DOI: 10.1038/s41598-025-94839-9] [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: 07/16/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Copy number variants (CNVs) are an important class of genomic variation known to be important for human physiology and diseases. Here we present genome-wide metabolomic signatures for CNVs in two Finnish cohorts-The Northern Finland Birth Cohort 1966 (NFBC 1966) and NFBC 1986. We have analysed and reported CNVs in over 9,300 individuals and characterised their dosage effect (CNV-metabolomic QTL) on 228 plasma lipoproteins and metabolites. We have reported reference (normal physiology) metabolomic signatures for up to ~ 2.6 million COVID-19 GWAS results from the National Institutes of Health (NIH) GRASP database, including for outcomes related to COVID-19 death, severity, and hospitalisation. Furthermore, by analysing two exemplar genes for COVID-19 severity namely LZTFL1 and OAS1, we have reported here two additional candidate genes for COVID-19 severity biology, (1) NFIX, a gene related to viral (adenovirus) replication and hematopoietic stem cells and (2) ACSL1, a known candidate gene for sepsis and bacterial inflammation. Based on our results and current literature we hypothesise that (1) charge imbalance across the cellular membrane between cations (Fe2+, Mg2+ etc.) and anions (e.g. ROS, hydroxide ion from cellular Fenton reactions, superoxide etc.), (2) iron trafficking within and between different cell types e.g., macrophages and (3) systemic oxidative stress response (e.g. lipid peroxidation mediated inflammation), together could be of relevance in severe COVID-19 cases. To conclude, our unique atlas of univariate and multivariate metabolomic signatures for CNVs (~ 7.2 million signatures) with deep annotations of various multi-omics data sets provide an important reference knowledge base for human metabolism and diseases.
Collapse
Affiliation(s)
- Tisham De
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
- Department of Genomics of Common Diseases, Imperial College London, London, UK.
- Department of Infectious Disease, Imperial College London, London, UK.
| | - Lachlan Coin
- Department of Infectious Disease, Imperial College London, London, UK
- Department of Microbiology and Immunology, Institute for Infection and Immunity, University of Melbourne at The Peter Doherty, Melbourne, Australia
| | - Jethro Herberg
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Marjo-Riitta Järvelin
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Centre for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care and Medical Research Center, Oulu University Hospital, Oulu, Finland
- Centre for Environment and Health, Imperial College London, London, UK
- Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
17
|
Randolph LN, Castiglioni C, Tavian M, Sturgeon CM, Ditadi A. Bloodhounds chasing the origin of blood cells. Trends Cell Biol 2025:S0962-8924(25)00067-4. [PMID: 40221343 DOI: 10.1016/j.tcb.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025]
Abstract
The generation of blood cells during embryonic development involves a process resembling lineage reprogramming, where specialized cells within the vasculature become blood forming, or hemogenic. These hemogenic cells undergo rapid transcriptional and morphological changes as they appear to switch from an endothelial to blood identity. What controls this process and the exact nature of the hemogenic cells remains debated, with evidence supporting several hypotheses. In this opinion, we synthesize current knowledge and propose a model reconciling conflicting observations, integrating evolutionary and mechanistic insights into blood cell emergence.
Collapse
Affiliation(s)
- Lauren N Randolph
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Castiglioni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Manuela Tavian
- University of Strasbourg, INSERM UMR-S1109, FMTS, Strasbourg, France
| | - Christopher M Sturgeon
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Ditadi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
18
|
Mo X, Shen A, Han Y, Xu L, Miao J, Xu D, Ji Q, Cao Y, Ge G, Zhu X, Deng H. Polysaccharide Nanoadjuvants Engineered via Phenotype-Specific Nanoprobe-Assisted Phenotypic Screen Reprogram Macrophage Cell Functions for Cancer and Rheumatoid Arthritis Therapy. ACS NANO 2025; 19:12920-12936. [PMID: 40152971 PMCID: PMC11984301 DOI: 10.1021/acsnano.4c16671] [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: 11/20/2024] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 03/30/2025]
Abstract
Macrophage plays critical roles in immune-related diseases, acting as a crucial therapeutic target for immunotherapy. Rational design and development of effective therapeutics for macrophage reprogramming are still challenging. Here, we rationally engineered polysaccharide nanoadjuvants to reprogram macrophage functions for enhanced immunotherapy in multiple diseases through a macrophage phenotype-specific nanoprobe (MPSNPr)-assisted high-throughput phenotypic screen. This MPSNPr exhibited high macrophage M1 phenotype specificity because of the formation of H-aggregates on the outer surface and the binding to glucose transporter 1 receptors by the polysaccharide nanocarrier. Based on this MPSNPr, a high-throughput platform was constructed and employed to screen a variety of pharmaceuticals for macrophage reprogramming, being able to identify both pro-inflammatory and anti-inflammatory drug candidates. Polysaccharide nanoadjuvants, Dex-BA and Dex-SAL, were rationally engineered with two potent candidates to amplify macrophage reprogramming efficacy both in vitro and in vivo. Dex-BA significantly inhibited tumor growth by inducing macrophage M1 polarization, dendritic cell maturation, and cytotoxic T cell activation in a mice melanoma model. Dex-SAL alleviated rheumatoid arthritis symptoms with reduced inflammation by reprogramming activated macrophages toward anti-inflammatory phenotype. Our work provides a robust strategy for the rational design and development of effective therapeutics for enhanced macrophage-mediated immunotherapy in diverse diseases.
Collapse
Affiliation(s)
- Xuan Mo
- Shanghai
Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary
Integrative Medicine Research, Shanghai
University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Anping Shen
- Shi’s
Center of Orthopedics and Traumatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yicun Han
- Department
of Medical Oncology & Cancer Institute of Integrative Medicine,
Shuguang Hospital, Shanghai University of
Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Xu
- Shanghai
Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary
Integrative Medicine Research, Shanghai
University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaqian Miao
- Shanghai
Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary
Integrative Medicine Research, Shanghai
University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Danni Xu
- Shanghai
Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary
Integrative Medicine Research, Shanghai
University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qing Ji
- Department
of Medical Oncology & Cancer Institute of Integrative Medicine,
Shuguang Hospital, Shanghai University of
Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuelong Cao
- Shi’s
Center of Orthopedics and Traumatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guangbo Ge
- Shanghai
Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary
Integrative Medicine Research, Shanghai
University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinyuan Zhu
- School
of Chemistry and Chemical Engineering, Frontiers Science Center for
Transformative Molecules, Shanghai Jiao
Tong University, Shanghai 200240, China
| | - Hongping Deng
- Shanghai
Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary
Integrative Medicine Research, Shanghai
University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
19
|
Wu W, Meng T, Wang Y, Chen J, Liang C. Prdx5 regulates macrophage polarization by modulating the TLR4/NF-κB pathway to promote apoptosis in chronic prostatitis. Int Immunopharmacol 2025; 151:114332. [PMID: 40015209 DOI: 10.1016/j.intimp.2025.114332] [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: 11/13/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 03/01/2025]
Abstract
Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a prevalent urological disorder characterized by urinary symptoms, pelvic pain, and sexual dysfunction. Despite its high prevalence, the pathogenesis of CP/CPPS remains poorly understood. Our study revealed that peroxiredoxin 5 (Prdx5) was upregulated in M1 macrophages and in mice with experimental autoimmune prostatitis (EAP), with its expression in macrophages being regulated in a reactive oxygen species (ROS)-dependent manner. Using western blotting, RT-qPCR, immunohistochemical staining, hematoxylin and eosin staining, immunofluorescence staining, flow cytometry, and cell co-culturing, it was demonstrated that the silencing of Prdx5 suppressed the polarization of macrophages towards the M1 phenotype. This inhibition reduced apoptosis in prostate epithelial cells and mitigated the progression of EAP. Furthermore, Prdx5 mediated its effects in macrophages and EAP via the Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) pathway. Our findings suggest that Prdx5 promoted the occurrence and development of CP/CPPS due to its promotion of M1 polarization and apoptosis of prostate epithelial cells in an ROS-dependent manner via the TLR4/NF-κB axis, indicating its potential as a therapeutic target to treat CP/CPPS.
Collapse
Affiliation(s)
- Weikang Wu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China
| | - Tong Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China
| | - Yufan Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China
| | - Jing Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China.
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China.
| |
Collapse
|
20
|
Li S, Zhou X, Duan Q, Niu S, Li P, Feng Y, Zhang Y, Xu X, Gong SP, Cao H. Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis. Int J Mol Sci 2025; 26:3252. [PMID: 40244103 PMCID: PMC11989900 DOI: 10.3390/ijms26073252] [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: 01/27/2025] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by lipid accumulation and immune cell infiltration, is linked to plaque formation and cardiovascular events. While traditionally associated with lipid metabolism and endothelial dysfunction, recent research highlights the roles of autophagy and clonal hematopoiesis (CH) in its pathogenesis. Autophagy, a cellular process crucial for degrading damaged components, regulates macrophage homeostasis and inflammation, both of which are pivotal in atherosclerosis. In macrophages, autophagy influences lipid metabolism, cytokine regulation, and oxidative stress, helping to prevent plaque instability. Defective autophagy exacerbates inflammation, impairs cholesterol efflux, and accelerates disease progression. Additionally, autophagic processes in endothelial cells and smooth muscle cells further contribute to atherosclerotic pathology. Recent studies also emphasize the interplay between autophagy and CH, wherein somatic mutations in genes like TET2, JAK2, and DNMT3A drive immune cell expansion and enhance inflammatory responses in atherosclerotic plaques. These mutations modify macrophage function, intensifying the inflammatory environment and accelerating atherosclerosis. Chaperone-mediated autophagy (CMA), a selective form of autophagy, also plays a critical role in regulating macrophage inflammation by degrading pro-inflammatory cytokines and oxidized low-density lipoprotein (ox-LDL). Impaired CMA activity leads to the accumulation of these substrates, activating the NLRP3 inflammasome and worsening inflammation. Preclinical studies suggest that pharmacologically activating CMA may mitigate atherosclerosis progression. In animal models, reduced CMA activity accelerates plaque instability and increases inflammation. This review highlights the importance of autophagic regulation in macrophages, focusing on its role in inflammation, plaque formation, and the contributions of CH. Building upon current advances, we propose a hypothesis in which autophagy, programmed cell death, and clonal hematopoiesis form a critical intrinsic axis that modulates the fundamental functions of macrophages, playing a complex role in the development of atherosclerosis. Understanding these mechanisms offers potential therapeutic strategies targeting autophagy and inflammation to reduce the burden of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Shuanhu Li
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
| | - Xin Zhou
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Qinchun Duan
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shukun Niu
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Pengquan Li
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Yihan Feng
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Ye Zhang
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Xuehong Xu
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shou-Ping Gong
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Huiling Cao
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| |
Collapse
|
21
|
Han Z, Shen Y, Yan Y, Bin P, Zhang M, Gan Z. Metabolic reprogramming shapes post-translational modification in macrophages. Mol Aspects Med 2025; 102:101338. [PMID: 39977975 DOI: 10.1016/j.mam.2025.101338] [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: 09/11/2024] [Revised: 12/10/2024] [Accepted: 01/11/2025] [Indexed: 02/22/2025]
Abstract
Polarized macrophages undergo metabolic reprogramming, as well as extensive epigenetic and post-translational modifications (PTMs) switch. Metabolic remodeling and dynamic changes of PTMs lead to timely macrophage response to infection or antigenic stimulation, as well as its transition from a pro-inflammatory to a reparative phenotype. The transformation of metabolites in the microenvironment also determines the PTMs of macrophages. Here we reviewed the current understanding of the altered metabolites of glucose, lipids and amino acids in macrophages shape signaling and metabolism pathway during macrophage polarization via PTMs, and how these metabolites in some macrophage-associated diseases affect disease progression by shaping macrophage PTMs.
Collapse
Affiliation(s)
- Ziyi Han
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yinhao Shen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuqi Yan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Peng Bin
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Meimei Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Zhending Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China.
| |
Collapse
|
22
|
Yao TX, Li N, Huang LS. Integrated single-cell transcriptomic map of pig kidney cells across various periods and anatomical sites. Zool Res 2025; 46:469-482. [PMID: 40116025 PMCID: PMC12000134 DOI: 10.24272/j.issn.2095-8137.2024.335] [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: 12/07/2024] [Accepted: 01/14/2025] [Indexed: 03/23/2025] Open
Abstract
The kidney is essential for maintaining fluid, electrolyte, and metabolite homeostasis, and for regulating blood pressure. The pig serves as a valuable biomedical model for human renal physiology, offering insights across different physiological states. In this study, single-cell RNA sequencing was used to profile 138 469 cells from 12 pig kidney samples collected during the embryonic (E), fattening (F), and pregnancy (P) periods, identifying 29 cell types. Proximal tubule (PT) cells exhibited elevated expression of metabolism-related transcription factors (TFs), including GPD1, ACAA1, and AGMAT, with validation across multiple individuals, periods, and species. Fluorescence homologous double-labeling of paraffin sections further confirmed the expression of ACAA1 and AGMAT in PT cells. Comparative analysis of pig and human kidneys revealed a high degree of similarity among corresponding cell types. Analysis of cell-type heterogeneity highlighted the diversity of thick ascending limb (TAL) cells, identifying a TAL subpopulation related to immune function. Additionally, the functional heterogeneity of kidney-resident macrophages (KRM) was explored across different anatomical sites. In the renal medulla, KRM were implicated in phagocytosis and leukocyte activation, whereas in the renal pelvis, they functioned as ligands, recruiting neutrophils with bactericidal activity to the renal pelvis to combat urinary tract infections.
Collapse
Affiliation(s)
- Tian-Xiong Yao
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China. E-mail:
| | - Na Li
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Lu-Sheng Huang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China. E-mail:
| |
Collapse
|
23
|
Stephan JK, Knerr T, Gu Z, Li H, Brittian KR, Dassanayaka S, Singhal R, Nong Y, Jones SP, Wysoczynski M. Neutrophil-secreted CHI3L1 exacerbates cardiac dysfunction and inflammation after myocardial infarction. FASEB J 2025; 39:e70422. [PMID: 40013912 PMCID: PMC11963974 DOI: 10.1096/fj.202401654r] [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: 07/17/2024] [Revised: 01/24/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Myocardial infarction (MI) triggers acute inflammation, marked by neutrophil infiltration. Although neutrophils are central to this response, the exact role of various neutrophil-derived factors is not fully understood. Clinical studies have linked one such enigmatic factor, chitinase-3 like-1, to MI outcomes. Hence, we investigated its role in post-MI remodeling. We found that chitinase-3 like-1 (CHI3L1) is upregulated after MI and secreted by activated neutrophils but does not directly affect neutrophil activity. To assess whether increased CHI3L1 influences ventricular remodeling, we subjected mice to non-reperfused MI and administered recombinant CHI3L1. Increased CHI3L1 levels worsened ventricular remodeling. In contrast, CHI3L1-deficient mice showed reduced ventricular remodeling after MI. To explore the underlying mechanisms, we assessed interactions with other cells known to be important in ventricular remodeling. Immunoprofiling of infarcted CHI3L1-deficient mouse hearts revealed a faster decline in neutrophil and monocyte numbers, indicating quicker resolution of inflammation. These findings provide direct evidence that CHI3L1 exacerbates ventricular inflammation and remodeling following MI through gain- and loss-of-function approaches.
Collapse
Affiliation(s)
- Jonah K. Stephan
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Taylor Knerr
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Zhen Gu
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Hong Li
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Kenneth R. Brittian
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Sujith Dassanayaka
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Richa Singhal
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Yibing Nong
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Steven P. Jones
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Marcin Wysoczynski
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| |
Collapse
|
24
|
Moraitis I, Taelman J, Arozamena B, Mularoni L, Wienskowska O, Sanjuan Garriga X, Arregui L, Stefanovic M, Modolell Farré I, Guedea F, Diaz M, Guiu J. Mucosal Macrophages Govern Intestinal Regeneration in Response to Injury. Gastroenterology 2025:S0016-5085(25)00465-2. [PMID: 40086603 DOI: 10.1053/j.gastro.2025.01.252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 01/15/2025] [Accepted: 01/30/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND & AIMS Radiation-induced enteritis develops in cancer patients treated with radiotherapy in the abdominal and pelvic cavity, a condition that impairs their quality of life. Radiation injury depletes proliferative intestinal stem cells; in response to this, the epithelium activates a regenerative program that facilitates the healing of the intestine. However, the mechanisms that induce the activation of the intestinal regenerative program are poorly characterized. METHODS In this study, we induced radiation-induced enteritis in mice through abdominal irradiation, mimicking clinical scenarios. Through imaging and flow cytometric analysis, we investigated the recruitment of macrophages to the small intestine during injury and healing. Additionally, we developed a coculture system for mouse and human intestinal organoids and macrophages to explore the cross talk between these cells. Then by combining in vivo ablation of macrophages, fluorescent lineage tracing, imaging, bulk RNA-sequencing (RNA-seq), single-cell RNA-seq, human intestinal organoids, and cell trajectory analysis, we studied the macrophage induction of intestinal regeneration at the cellular and molecular level. RESULTS Our findings revealed that macrophages are recruited around the intestinal stem cell compartment upon radiation injury, promoting a fetal-like reprogramming and proliferation of epithelial cells that drives the regeneration process. In contrast, macrophage ablation led to compromised regeneration. Moreover, our single-cell RNA-seq analysis identified key secreted molecules, neuregulin 1 and osteopontin, as pivotal players in regulating this process. Additionally, characterization of human macrophage/organoid cocultures and cell trajectory inference confirmed the conservation of macrophages' role in triggering the regenerative program in primary human cells. CONCLUSIONS This study identifies macrophages as essential contributors to intestinal regeneration beyond their innate immune response. Targeting macrophages therapeutically may hold promise in enhancing regeneration and improving the quality of life for cancer survivors.
Collapse
Affiliation(s)
- Ilias Moraitis
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain; Biomedicine PhD Program, Universitat de Barcelona, Barcelona, Spain
| | - Jasin Taelman
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Borja Arozamena
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Loris Mularoni
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Olga Wienskowska
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Xavier Sanjuan Garriga
- Department of Pathology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Laura Arregui
- HUB-ICO-IDIBELL Biobank, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Milica Stefanovic
- Department of Radiobiology and Cancer, ONCOBELL, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Department of Radiation Oncology, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ignasi Modolell Farré
- Servei de Física Mèdica i Protecció Radiològica, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ferran Guedea
- Department of Radiobiology and Cancer, ONCOBELL, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Department of Radiation Oncology, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mònica Diaz
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Guiu
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain; Centre for Networked Biomedical Research on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
25
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
26
|
Shin SD, Branning R, McGinnis M, Shin A, Ho M, Karpyak VM, Oesterle T. Buprenorphine's Effect on the Human Immune System and Inflammation. Clin Transl Sci 2025; 18:e70180. [PMID: 40065582 PMCID: PMC11893730 DOI: 10.1111/cts.70180] [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: 01/22/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Opioid use disorder is a persistent epidemic despite several FDA-approved medications for its treatment. While the pathogenesis of opioid use disorder has been classically attributed to dopamine pathways in the brain, there is emerging evidence and interest surrounding the role of inflammation and inflammatory signaling in its development and treatment. Buprenorphine has become the most prescribed medication for opioid use disorder, largely due to its ease of access and tolerability. This review aimed to better characterize contemporary knowledge of how buprenorphine modulates the human immune system and inflammatory functions in this population. A comprehensive review was conducted using 11 key databases, including Embase, MEDLINE, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov. This review captured 8177 records, and 14 studies were ultimately selected for inclusion and discussion in this review. Notably, all 14 clinical studies evaluated buprenorphine's effect on the peripheral immune system, and the majority of the studies supported the notion that initiation and maintenance of buprenorphine restore immune suppression caused by opioid use disorder. In addition, we discuss how recent and ongoing work utilizing advanced imaging and cellular technologies is advancing the understanding of how buprenorphine affects the immune and inflammatory signaling in the brain.
Collapse
Affiliation(s)
- Samuel D. Shin
- Department of Psychiatry and PsychologyCleveland ClinicClevelandOhioUSA
| | - Rachel Branning
- Department of PsychiatryUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
| | | | - Alexandra Shin
- Department of Pathology and Human AnatomyLoma Linda UniversityLoma LindaCaliforniaUSA
| | - Ming‐Fen Ho
- Department of Psychiatry & PsychologyMayo ClinicRochesterMinnesotaUSA
- Department of Molecular Pharmacology and Experimental TherapeuticsMayo ClinicRochesterMinnesotaUSA
| | - Victor M. Karpyak
- Department of Psychiatry & PsychologyMayo ClinicRochesterMinnesotaUSA
| | - Tyler Oesterle
- Department of Psychiatry & PsychologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
27
|
Zeng Y, Yuan W, Feng C, Peng L, Xie X, Peng F, Li T, Lin M, Zhang H, Dai H. Trametinib alleviates lipopolysaccharide-induced acute kidney injury by inhibiting macrophage polarization through the PI3K/Akt pathway. Transpl Immunol 2025; 89:102183. [PMID: 39892762 DOI: 10.1016/j.trim.2025.102183] [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: 12/25/2024] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Sepsis-induced acute kidney injury (AKI) is a severe condition characterized by dysregulation of pro- and anti-inflammatory responses. Targeting macrophage polarization between pro-inflammatory M1 and anti-inflammatory M2 cells offers a potential therapeutic approach for AKI. Trametinib (TRAM), an inhibitor of the MEK1/2 signaling pathway, was evaluated for its impact on M1/M2 polarization in AKI. METHODS Wild-type (WT) mice were subjected to lipopolysaccharide (LPS)-induced AKI and intraperitoneally treated with dimethyl sulfoxide (DMSO) or TRAM (10 mg/kg) for three days. Renal function was assessed by measuring creatinine levels. While histopathological changes, RNA sequencing data, and serum cytokine levels were analyzed. Macrophage M1/M2 polarization in kidney tissues was examined using flow cytometry and immunohistochemistry. Murine bone marrow-derived macrophages (BMDMs) were polarized to the M1 or M2 phenotype in vivo and treated with or without TRAM (10 μM). M1/M2 polarization was analyzed via flow cytometry, and PI3K/Akt signaling was evaluated by western blotting. RESULTS TRAM significantly improved renal function, as demonstrated by reduced serum creatinine levels (p < 0.01) and ameliorated histopathological damage (p < 0.01). Flow cytometry and immunohistochemistry revealed that TRAM markedly inhibited pro-inflammatory M1 macrophage polarization (p < 0.001). Additionally, TRAM reduced serum level of IFN-γ (p < 0.01) and IL-17 (p < 0.001). In vitro, TRAM suppressed M1 polarization (p < 0.05) by inhibiting the PI3K/Akt signaling pathway. CONCLUSION TRAM mitigated LPS-induced AKI by suppressing M1 macrophage polarization via the PI3K/Akt pathway, highlighting its therapeutic potential for AKI and other inflammatory kidney diseases.
Collapse
Affiliation(s)
- Yingqi Zeng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Wenjia Yuan
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Chen Feng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Longkai Peng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Xubiao Xie
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Fenghua Peng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Tengfang Li
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Minjie Lin
- Academic Affairs Department, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Hedong Zhang
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China.
| | - Helong Dai
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China.
| |
Collapse
|
28
|
Hooda V, Sharma A. Interactions of NK Cells and Macrophages: From Infections to Cancer Therapeutics. Immunology 2025; 174:287-295. [PMID: 39739619 DOI: 10.1111/imm.13886] [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: 08/25/2024] [Revised: 11/13/2024] [Accepted: 12/07/2024] [Indexed: 01/02/2025] Open
Abstract
The interaction between immune cells brings a consequence either on their role and functioning or the functioning of the other immune cells, modulating the whole mechanistic pathway. The interaction between natural killer (NK) cells and macrophages is one such interaction which is relatively less explored amongst diseased conditions. Their significance comes from their innate nature and secretion of large proportions of cytokines and chemokines which results in influencing adaptive immune responses. Their interplay can lead to several functional outcomes such as NK cell activation/inhibition, increased cytotoxicity and IFNγ release by NK cells, inhibition of macrophage function, etc. This paper delves into the interaction amongst NK cells and macrophages via different receptor-ligands and cytokines, particularly emphasising microbial infections and tumours. The review has the potential to uncover new insights and approaches that could lead to the development of innovative therapeutic tools and targets.
Collapse
Affiliation(s)
- Vishakha Hooda
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
29
|
Joshi JC, Joshi B, Zhang C, Banerjee S, Vellingiri V, Raghunathrao VAB, Anwar M, Rokade TP, Zhang L, Amin R, Song Y, Mehta D. RGS2 is an innate immune checkpoint for suppressing Gαq-mediated IFNγ generation and lung injury. iScience 2025; 28:111878. [PMID: 40041768 PMCID: PMC11876898 DOI: 10.1016/j.isci.2025.111878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 03/01/2024] [Accepted: 01/20/2025] [Indexed: 03/12/2025] Open
Abstract
Interferon gamma (IFNγ), a type II interferon, augments tissue inflammation following infections, leading to lethal acute lung injury (ALI), yet the mechanisms controlling IFNγ generation in the lungs remain elusive. Here, we identified regulator of G protein signaling 2 (RGS2) as a gatekeeper of the lung's IFNγ levels during infections. Deletion of RGS2 sustained an increase in IFNγ levels in macrophages, leading to unresolvable inflammatory lung injury. This response was not seen in RGS2 null chimeric mice receiving wild-type (WT) bone marrow or the RGS2 gene in alveolar macrophages (AMs) or IFNγ-blocking antibody. RGS2 functioned by suppressing Gαq-mediated IFNγ generation and AM inflammatory signaling. Thus, the inhibition of Gαq blocked IFNγ generation in AMs and rewired AM transcriptomes from an inflammatory to a reparative phenotype in RGS2 null mice, pointing to the RGS2-Gαq axis as a potential target for suppressing inflammatory injury.
Collapse
Affiliation(s)
- Jagdish Chandra Joshi
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
- Lake Erie College of Osteopathic Medicine, School of Pharmacy, Erie, PA, USA
| | - Bhagwati Joshi
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Cuiping Zhang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Somenath Banerjee
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Vigneshwaran Vellingiri
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Vijay Avin Balaji Raghunathrao
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Mumtaz Anwar
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Tejas Pravin Rokade
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Lianghui Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Vascular Medicine Institute, Center for Vaccine Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Ruhul Amin
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dolly Mehta
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| |
Collapse
|
30
|
Prado C, Herrada AA, Hevia D, Goiry LG, Escobedo N. Role of innate immune cells in multiple sclerosis. Front Immunol 2025; 16:1540263. [PMID: 40034690 PMCID: PMC11872933 DOI: 10.3389/fimmu.2025.1540263] [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: 12/05/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune, inflammatory and neurodegenerative disease affecting the central nervous system (CNS). MS is associated with a complex interplay between neurodegenerative and inflammatory processes, mostly attributed to pathogenic T and B cells. However, a growing body of preclinical and clinical evidence indicates that innate immunity plays a crucial role in MS promotion and progression. Accordingly, preclinical and clinical studies targeting different innate immune cells to control MS are currently under study, highlighting the importance of innate immunity in this pathology. Here, we reviewed recent findings regarding the role played by innate immune cells in the pathogenesis of MS. Additionally, we discuss potential new treatments for MS based on targets against innate immune components.
Collapse
Affiliation(s)
- Carolina Prado
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Andrés A. Herrada
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Daniel Hevia
- Center for Studies and Innovation in Dentistry, Facultad de Odontología, Universidad Finis Terrae, Santiago, Chile
| | - Lorna Galleguillos Goiry
- Neurology and Psychiatry Department, Clínica Alemana, Neurology and Neurosurgery Department, Clínica Dávila, Santiago, Chile
| | - Noelia Escobedo
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
31
|
Rayasam A, Moe A, Kudek M, Shah RK, Yuan CY, Miller JM, Rau M, Patton M, Wanat K, Colonna M, Zamora AE, Drobyski WR. Intestinal epithelium-derived IL-34 reprograms macrophages to mitigate gastrointestinal tract graft-versus-host disease. Sci Transl Med 2025; 17:eadn3963. [PMID: 39937882 DOI: 10.1126/scitranslmed.adn3963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 10/10/2024] [Accepted: 01/08/2025] [Indexed: 02/14/2025]
Abstract
Gastrointestinal (GI) tract graft-versus-host disease (GVHD) is a major complication after allogeneic hematopoietic stem cell transplantation and is attributable to dysregulation that occurs between the effector and regulatory arms of the immune system. Whereas regulatory T cells have a primary role in counterbalancing GVHD-induced inflammation, identifying and harnessing other pathways that promote immune tolerance remain major goals in this disease. Herein, we identified interleukin-34 (IL-34) as an intestinal epithelium-derived cytokine that was able to mitigate the severity of GVHD within the GI tract. Specifically, we observed that the absence of recipient IL-34 production exacerbated GVHD lethality, promoted intestinal epithelial cell death, and compromised barrier integrity. Mechanistically, the absence of host IL-34 skewed donor macrophages toward a proinflammatory phenotype and augmented the accumulation of pathogenic CD4+ granulocyte-macrophage colony-stimulating factor (GM-CSF)+ T cells within the colon. Conversely, the administration of recombinant IL-34 substantially reduced GVHD mortality and inflammation, which was dependent on the expression of apolipoprotein E in donor macrophages. Complementary genetic and imaging approaches in mice demonstrated that intestinal epithelial cells were the relevant source of IL-34. These results were supported by colonic biopsies from patients with GVHD, which displayed IL-34 expression in intestinal epithelial cells and apolipoprotein E in lamina propria macrophages, validating similar cellular localization in humans. These studies indicate that IL-34 acts as a tissue-intrinsic cytokine that regulates GVHD severity in the GI tract and could serve as a potential therapeutic target for amelioration of this disease.
Collapse
Affiliation(s)
- Aditya Rayasam
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Alison Moe
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew Kudek
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ravi K Shah
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cheng-Yin Yuan
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - James M Miller
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mary Rau
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mollie Patton
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Karolyn Wanat
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University, Saint Louis, MO 63110, USA
| | - Anthony E Zamora
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - William R Drobyski
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
32
|
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
|
33
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [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: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
34
|
Yi Y, Wu MY, Chen KT, Chen AH, Li LQ, Xiong Q, Wang XR, Lei WB, Xiong GX, Fang SB. LDHA-mediated glycolysis in stria vascularis endothelial cells regulates macrophages function through CX3CL1-CX3CR1 pathway in noise-induced oxidative stress. Cell Death Dis 2025; 16:65. [PMID: 39900910 PMCID: PMC11791080 DOI: 10.1038/s41419-025-07394-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 01/06/2025] [Accepted: 01/24/2025] [Indexed: 02/05/2025]
Abstract
According to the World Health Organization, more than 12% of the world's population suffers from noise-induced hearing loss (NIHL). Oxidative stress-mediated damage to the stria vascularis (SV) is one of the pathogenic mechanisms of NIHL. Recent studies indicate that glycolysis plays a critical role in endothelial cells (ECs)-related diseases. However, the specific role of glycolysis in dysfunction of SV-ECs remain largely unknown. In this study, we investigated the effects of glycolysis on SV-ECs in vitro and on the SV in vivo. Our previous research identified the glycolysis pathway as a potential mechanism underlying the SV-ECs injuries induced by oxidative stress. We further examined the expression levels of glycolytic genes in SV-ECs under H2O2 stimulation and in noise-exposed mice. We found that the gene and protein expression levels of glycolytic-related enzyme LDHA significantly decreased at early phase after oxidative stress injury both in vitro and in vivo, and exhibited anti-inflammatory effects on macrophages (Mφ). Moreover, we analyzed the differential secretomes of SV-ECs with and without inhibition of LDHA using LC-MS/MS technology, identifying CX3CL1 as a candidate mediator for cellular communication between SV-ECs and Mφ. We found that CX3CL1 secretion from SV-ECs was decreased following LDHA inhibition and exhibited anti-inflammatory effects on Mφ via the CX3CR1 pathway. Similarly, the pro-inflammatory effect of LDHA-overexpressing SV-ECs was attenuated following inhibition of CX3CL1. In conclusion, our study revealed that glycolysis-related LDHA was reduced in oxidative stress-induced SV-ECs, and that LDHA inhibition in SV-ECs elicited anti-inflammatory effects on Mφ, at least partially through the CX3CL1-CX3CR1 pathway. These findings suggest that LDHA represent a novel therapeutic strategy for the treatment of NIHL.
Collapse
Affiliation(s)
- Ying Yi
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China
| | - Min-Yu Wu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China
| | - Kai-Tian Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China
| | - An-Hai Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China
| | - Lin-Qiu Li
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China
| | - Qin Xiong
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China
| | - Xian-Ren Wang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China
| | - Wen-Bin Lei
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China.
| | - Guan-Xia Xiong
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China.
| | - Shu-Bin Fang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
35
|
Dong X, Xu H, He B, Zhang M, Miu W, Huang Z, Chen C. FIBRINOGEN-LIKE PROTEIN 2 PROTECTS THE AGGRAVATION OF HYPERTRIGLYCERIDEMIA ON THE SEVERITY OF HYPERTRIGLYCERIDEMIA ACUTE PANCREATITIS BY REGULATING MACROPHAGES. Shock 2025; 63:327-337. [PMID: 39527492 DOI: 10.1097/shk.0000000000002503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
ABSTRACT Objective: The mechanisms underlying the increased severity of hypertriglyceridemia acute pancreatitis (HTG-AP) remain poorly understood. Fibrinogen-like protein 2 (FGL2) has been identified as a regulator of macrophage activity, mediating immune suppression. This study aims to examine the role of FGL2 in the susceptibility to severe conditions of HTG-AP. Methods: Both wild-type and FGL2 gene knockout C57BL/6 mice were utilized to establish HTG, AP, and HTG-AP models using P-407 and/or caerulein. Serum levels of triglycerides, total cholesterol, amylase, and lipase were assessed via biochemical analysis. Pancreatic and lung tissue injuries were evaluated using hematoxylin and eosin staining. TNF-α, IL-1β, and IL-6 levels in serum and pancreatic tissues were quantified using enzyme-linked immunosorbent assay. Immunohistochemistry was used to assess the expression of FGL2, the macrophage marker CD68, and M1/M2 macrophage markers iNOS/CD163. Results: The animal models were successfully established. Compared to wild-type mice, FGL2 knockout resulted in increased pathological injury scores in the pancreas and lungs, as well as elevated TNF-α, IL-1β, and IL-6 levels in serum and pancreatic tissue in the HTG group, with more pronounced effects observed in the HTG-AP group. The AP group alone did not exhibit significant changes due to FGL2 knockout. Further analysis revealed that FGL2 knockout increased CD68 expression but reduced CD163 expression in the pancreatic tissues in the HTG group. In the HTG-AP group, there was a marked increase in CD68 and iNOS expressions, coupled with a reduction in CD163 expression. Conclusion: FGL2 knockout in HTG and HTG-AP mice resulted in increased inflammatory responses and a significant imbalance in M2 macrophages. These findings suggest that FGL2 plays a crucial role in mitigating the aggravation of HTG on the severity of HTG-AP by modulating macrophage activity.
Collapse
Affiliation(s)
- Xiuli Dong
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Haibo Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Interventional Pulmonology of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Baiqi He
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Interventional Pulmonology of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Meijuan Zhang
- Department of Medical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Wanqi Miu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Interventional Pulmonology of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Zhiming Huang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chengshui Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Interventional Pulmonology of Zhejiang Province, Wenzhou, Zhejiang Province, China
| |
Collapse
|
36
|
Park J, Kim D. Advanced Immunomodulatory Biomaterials for Therapeutic Applications. Adv Healthc Mater 2025; 14:e2304496. [PMID: 38716543 PMCID: PMC11834384 DOI: 10.1002/adhm.202304496] [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: 12/16/2023] [Revised: 04/15/2024] [Indexed: 05/22/2024]
Abstract
The multifaceted biological defense system modulating complex immune responses against pathogens and foreign materials plays a critical role in tissue homeostasis and disease progression. Recently developed biomaterials that can specifically regulate immune responses, nanoparticles, graphene, and functional hydrogels have contributed to the advancement of tissue engineering as well as disease treatment. The interaction between innate and adaptive immunity, collectively determining immune responses, can be regulated by mechanobiological recognition and adaptation of immune cells to the extracellular microenvironment. Therefore, applying immunomodulation to tissue regeneration and cancer therapy involves manipulating the properties of biomaterials by tailoring their composition in the context of the immune system. This review provides a comprehensive overview of how the physicochemical attributes of biomaterials determine immune responses, focusing on the physical properties that influence innate and adaptive immunity. This review also underscores the critical aspect of biomaterial-based immune engineering for the development of novel therapeutics and emphasizes the importance of understanding the biomaterials-mediated immunological mechanisms and their role in modulating the immune system.
Collapse
Affiliation(s)
- Ji‐Eun Park
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Dong‐Hwee Kim
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- Department of Integrative Energy EngineeringCollege of EngineeringKorea UniversitySeoul02841Republic of Korea
- Biomedical Research CenterKorea Institute of Science and TechnologySeoul02792Republic of Korea
| |
Collapse
|
37
|
Chen Y, Luo X, Li Y, Liu L, Liu Z, Tan Y, Chen Y. PTX3 impairs granulosa cell function by promoting the secretion of inflammatory cytokines in M1 macrophages via the JAK pathway. Steroids 2025; 214:109541. [PMID: 39577792 DOI: 10.1016/j.steroids.2024.109541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Polycystic Ovary Syndrome (PCOS) is an endocrine disorder syndrome among women in their reproductive years and is often linked to chronic inflammation. Pentraxin 3 (PTX3), a member of the pentraxin protein family, plays a key role in inflammation. In our study, we explored whether PTX3 influences granulosa cell function via its involvement in inflammation. Our analysis revealed elevated PTX3 concentrations in the follicular fluid and granulosa cells of patients with PCOS. Overexpression of PTX3 promoted apoptosis in the cultured murine granulosa cell line KK1 and inhibited the proliferation of these cells. Additionally, it suppressed the expression of luteinizing hormone receptor (LHR) and follicle-stimulating hormone receptor (FSHR), as well as those of key enzymes involved in steroid hormone synthesis, CYP19A1, and HSD3β, leading to reduced secretion of estradiol and progesterone. Moreover, both recombinant PTX3 protein and PTX3 secreted by granulosa cells (GCs) promoted the secretion of inflammatory cytokines IL-1β, IL-6, and TNF-α by M1 macrophages via the JAK pathway, which impaired the function of granulosa cells. This study may advance the understanding of cell-cell interactions in follicles and the inflammatory factors that contribute to PCOS pathophysiology.
Collapse
Affiliation(s)
- Ying Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaofang Luo
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yizhe Li
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Linhong Liu
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhen Liu
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunyue Tan
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
38
|
Qu F, Xu B, Kang H, Wang H, Ji J, Pang L, Wu Y, Zhou Z. The role of macrophage polarization in ulcerative colitis and its treatment. Microb Pathog 2025; 199:107227. [PMID: 39675441 DOI: 10.1016/j.micpath.2024.107227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/27/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Macrophages have great plasticity. Typically, there are two of activated macrophages: M1 macrophages and M2 macrophages. Of them, M1 macrophages play a major role in responses that are pro-inflammatory, while M2 macrophages play an important part in responses that are anti-inflammatory. Ulcerative colitis (UC) is a chronic, non-specific inflammatory disease of the intestine. The pathophysiology and course of UC are significantly influenced by the inflammatory response triggered by macrophage activation. M1 is a possible cause of increased inflammation in UC whereas M2 has a significant function in the healing of inflammation. The polarization imbalance of intestinal M1/M2 macrophages is closely linked to UC. Thus, by suppressing M1 polarization, encouraging M2 polarization, and reestablishing macrophage polarization balance, the treatment of UC based on macrophage polarization is beneficial for UC. Not only chemical drugs, but also traditional Chinese medicine compounds and herbal extracts have been shown to restore the balance of macrophage polarization, providing a new idea in the treatment of UC.
Collapse
Affiliation(s)
- Fanfan Qu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Baoqing Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongchang Kang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Hongxia Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jianbin Ji
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lianjing Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yaqian Wu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhenghua Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
39
|
Mesquita T, Miguel-Dos-Santos R, Liu W, Fournier M, Rogers RG, Alfaro J, Nawaz A, Sanchez L, Jones XM, Li L, Marbán E, Cingolani E. Upregulated FoxO1 promotes arrhythmogenesis in mice with heart failure and preserved ejection fraction. Nat Commun 2025; 16:1184. [PMID: 39885127 PMCID: PMC11782541 DOI: 10.1038/s41467-025-56186-1] [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: 01/08/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
Myocardial fibrosis leads to cardiac dysfunction and arrhythmias in heart failure with preserved ejection fraction (HFpEF), but the underlying mechanisms remain poorly understood. Here, RNA sequencing identifies Forkhead Box1 (FoxO1) signaling as abnormal in male HFpEF hearts. Genetic suppression of FoxO1 alters the intercellular communication between cardiomyocytes and fibroblasts, alleviates abnormal diastolic relaxation, and reduces arrhythmias. Targeted downregulation of FoxO1 in activated fibroblasts reduces cardiac fibrosis, blunts arrhythmogenesis and improves diastolic function in HFpEF. These results not only implicate FoxO1 in arrhythmogenesis and lusitropy but also demonstrate that pro-fibrotic remodeling and cardiomyocyte-fibroblast communication can be corrected, constituting an alternative therapeutic strategy for HFpEF.
Collapse
Affiliation(s)
- Thassio Mesquita
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | | | - Weixin Liu
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Mario Fournier
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Russell G Rogers
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Jocelyn Alfaro
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Asma Nawaz
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Lizbeth Sanchez
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Xaviar M Jones
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Liang Li
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Eugenio Cingolani
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA.
| |
Collapse
|
40
|
Gu W, Zhao J, Xu Y. Hyperuricemia-induced complications: dysfunctional macrophages serve as a potential bridge. Front Immunol 2025; 16:1512093. [PMID: 39935474 PMCID: PMC11810932 DOI: 10.3389/fimmu.2025.1512093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
With the changes in modern life, hyperuricemia (HUA) has become a serious universal health issue, leading to rising morbidity and mortality. Characterized by elevated levels of UA, HUA has become an independent risk factor for gout, chronic kidney disease, insulin resistance, cardiovascular disease, nonalcoholic fatty liver disease, etc. As HUA is a metabolic syndrome, the immune response is likely to play an active role throughout the whole process. Moreover, macrophages, as an indispensable component of the immune system, may serve as a promising target for addressing hyperuricemia-induced inflammation. Along with their precursor cells, monocytes, macrophages play a key role in the pathogenesis of HUA, primarily through three specific aspects, all of which are associated with inflammatory cytokines. The first mechanism involves direct action on urate transporters, such as URAT1 and ABCG2. The second mechanism is the modulation of inflammation, including targeting toll-like receptors (TLRs) and the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome. The third mechanism pertains to the effects on oxidative stress mediators. In this review, we summarize the underlying mechanisms of hyperuricemia, focusing on the effects of macrophages, therapeutic approaches, and clinical trials addressing hyperuricemia-caused dysfunction. Additionally, we highlight directions for future development, aiming to support future theoretical studies.
Collapse
Affiliation(s)
- Wenyi Gu
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiajing Zhao
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Yu Xu
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for Traditional Chinese Medicine New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
41
|
Kundu S, Pal K, Pyne A, Wang X. Force-bearing phagocytic adhesion rings mediate the phagocytosis of surface-bound particles. Nat Commun 2025; 16:984. [PMID: 39856073 PMCID: PMC11759950 DOI: 10.1038/s41467-025-56404-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Many micro-particles including pathogens strongly adhere to hosts. It remains elusive how macrophages detach these surface-bound particles during phagocytosis. We show that, rather than binding directly to these particles, macrophages form unique β2 integrin-mediated adhesion structures at the cell-substrate interfaces, specifically encircling the surface-bound particles. These circular adhesion structures that we named phagocytic adhesion rings (PARs) serve as strongholds to support local ring-shaped actin structures constricting into the particle-substrate cleavages, thereby pinching off the particles from the substrate. During this process, integrins in PARs sustain tensions due to the reaction force of actin polymerization against the particles. Such tensions are critical for phagocytic efficiency of surface-bound particles. PARs were formed in all tested macrophages (mouse, human and fish) and micron-sized particles (microbeads and E. coli), demonstrating their conserved role in the phagocytosis. This study reveals a mechanism of PAR-mediated phagocytosis, specialized for the detachment and internalization of surface-bound particles.
Collapse
Affiliation(s)
- Subhankar Kundu
- Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Kaushik Pal
- Department of Chemistry, Indian Institute of Technology Tirupati, Yerpedu, India
| | - Arghajit Pyne
- Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Xuefeng Wang
- Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
42
|
Frenis K, Badalamenti B, Mamigonian O, Weng C, Wang D, Fierstein S, Côté P, Khong H, Li H, Lummertz da Rocha E, Sankaran VG, Rowe RG. Path of differentiation defines human macrophage identity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634694. [PMID: 39896569 PMCID: PMC11785145 DOI: 10.1101/2025.01.24.634694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Macrophages play central roles in immunity, wound healing, and homeostasis - a functional diversity that is underpinned by varying developmental origins. The impact of ontogeny on properties of human macrophages is inadequately understood. We demonstrate that definitive human fetal liver (HFL) hematopoietic stem cells (HSCs) possess two divergent paths of macrophage specification that lead to distinct identities. The monocyte-dependent pathway exists in both prenatal and postnatal hematopoiesis and generates macrophages with adult-like responses properties. We now uncover a fetal-specific pathway of expedited differentiation that generates tissue resident-like macrophages (TRMs) that retain HSC-like self-renewal programs governed by the aryl hydrocarbon receptor (AHR). We show that AHR antagonism promotes TRM expansion and mitigates inflammation in models of atopic dermatitis (AD). Overall, we directly connect path of differentiation with functional properties of macrophages and identify an approach to promote selective expansion of TRMs with direct relevance to inflammation and diseases of macrophage dysfunction.
Collapse
|
43
|
Li J, Xiao C, Li C, He J. Tissue-resident immune cells: from defining characteristics to roles in diseases. Signal Transduct Target Ther 2025; 10:12. [PMID: 39820040 PMCID: PMC11755756 DOI: 10.1038/s41392-024-02050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025] Open
Abstract
Tissue-resident immune cells (TRICs) are a highly heterogeneous and plastic subpopulation of immune cells that reside in lymphoid or peripheral tissues without recirculation. These cells are endowed with notably distinct capabilities, setting them apart from their circulating leukocyte counterparts. Many studies demonstrate their complex roles in both health and disease, involving the regulation of homeostasis, protection, and destruction. The advancement of tissue-resolution technologies, such as single-cell sequencing and spatiotemporal omics, provides deeper insights into the cell morphology, characteristic markers, and dynamic transcriptional profiles of TRICs. Currently, the reported TRIC population includes tissue-resident T cells, tissue-resident memory B (BRM) cells, tissue-resident innate lymphocytes, tissue-resident macrophages, tissue-resident neutrophils (TRNs), and tissue-resident mast cells, but unignorably the existence of TRNs is controversial. Previous studies focus on one of them in specific tissues or diseases, however, the origins, developmental trajectories, and intercellular cross-talks of every TRIC type are not fully summarized. In addition, a systemic overview of TRICs in disease progression and the development of parallel therapeutic strategies is lacking. Here, we describe the development and function characteristics of all TRIC types and their major roles in health and diseases. We shed light on how to harness TRICs to offer new therapeutic targets and present burning questions in this field.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
44
|
Han L, Wu T, Zhang Q, Qi A, Zhou X. Immune Tolerance Regulation Is Critical to Immune Homeostasis. J Immunol Res 2025; 2025:5006201. [PMID: 39950084 PMCID: PMC11824399 DOI: 10.1155/jimr/5006201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/07/2024] [Indexed: 02/16/2025] Open
Abstract
The body's immune response plays a critical role in defending against external or foreign antigens while also preserving tolerance to self-antigens. This equilibrium, referred to as immune homeostasis, is paramount for overall health. The regulatory mechanisms governing the maintenance of this delicate immune balance are notably complex. It is currently accepted that immune tolerance is a dynamic outcome regulated by multiple factors, including central and peripheral mechanisms. Its induction or elimination plays a significant role in autoimmune diseases, organ transplantation, and cancer therapy, markedly impacting various major diseases in modern clinical practice. Overall, our current understanding of immune tolerance is still very limited. In this review article, we summarized the main mechanisms that have been known to mediate immune tolerance so far, including endogenous immune tolerance, adaptive immune tolerance, other immune tolerance mechanisms, and the homeostasis of immune tolerance, identified the key factors that regulate immune tolerance, and provided new clues for immune system recovery in many autoimmune diseases, organ transplantation, and tumor therapy.
Collapse
Affiliation(s)
- Lei Han
- Department of Pharmacy, Jiangsu Health Vocational College, Nanjing 211800, Jiangsu, China
| | - Tianxiang Wu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Qin Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Anning Qi
- Medical Laboratory, Liuhe People's Hospital of Jiangsu Province, Nanjing, Jiangsu 211500, China
| | - Xiaohui Zhou
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
45
|
Hai Q, Bazeley P, Han J, Brubaker G, Powers J, Diaz-Montero CM, Smith JD. Optimized Method to Generate Well-Characterized Macrophages from Induced Pluripotent Stem Cells. Biomedicines 2025; 13:99. [PMID: 39857683 PMCID: PMC11762477 DOI: 10.3390/biomedicines13010099] [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: 12/20/2024] [Revised: 12/30/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Macrophages play a pivotal role in various pathogenic processes, necessitating the development of efficient differentiation techniques to meet the high demand for these cells in research and therapy. Human macrophages can be obtained via culturing peripheral blood monocytes; however, this source has limited yields and requires patient contact for each proposed use. In addition, it would be difficult to perform gene editing on peripheral blood monocytes. The objectives of this study are to define a robust and consistent method for the differentiation of induced pluripotent stem cells (iPSCs) into macrophages that can address these needs for recurrent studies with high yields and the potential for gene editing. Methods: We refined the traditional embryoid body-based differentiation strategy to create a novel three-phase method that optimizes yield, consistent quality, and reproducibility. This approach incorporates microwell plates and cell filtration to standardize the production of embryoid bodies and subsequent macrophage progenitors. Using up to five independent iPSC donors, we performed several assays for macrophage functions and polarization, such as marker protein staining by flow cytometry, lipoprotein uptake, phagocytosis, cytokine release, inflammasome activation, and the effects of M1-like and M2-like polarization. RNA sequencing was performed to determine the segregation of cells at different stages of differentiation and by iPSC donor, as well as to identify marker genes for each stage of differentiation. Results: The iPSC-derived macrophages generated through this method exhibit characteristic features and cell marker proteins, as well as classical macrophage activities, including lipoprotein uptake, bacterial phagocytosis, cytokine release, and inflammasome activation. We demonstrate the effects of M1-like and M2-like polarization on cytokine release. The first three principal components of the RNA sequencing data showed clear clustering by differentiation stage. In contrast, the fourth and fifth principal components clustered the differentiated macrophages by their respective iPSC donor. Marker genes were identified for each stage of differentiation and polarization. Conclusions: The methods provide an optimized and simplified procedure to produce iPSC-derived macrophages. Our results demonstrate the reproducibility of this method in generating high-quality macrophages suitable for a variety of biomedical applications.
Collapse
Affiliation(s)
- Qimin Hai
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Peter Bazeley
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Juying Han
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gregory Brubaker
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jennifer Powers
- Immunomonitoring Laboratory, Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Claudia M. Diaz-Montero
- Immunomonitoring Laboratory, Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jonathan D. Smith
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland, OH 44195, USA
| |
Collapse
|
46
|
Yan Y, Antolin N, Zhou L, Xu L, Vargas IL, Gomez CD, Kong G, Palmisano I, Yang Y, Chadwick J, Müller F, Bull AMJ, Lo Celso C, Primiano G, Servidei S, Perrier JF, Bellardita C, Di Giovanni S. Macrophages excite muscle spindles with glutamate to bolster locomotion. Nature 2025; 637:698-707. [PMID: 39633045 PMCID: PMC11735391 DOI: 10.1038/s41586-024-08272-5] [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: 10/07/2023] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
The stretch reflex is a fundamental component of the motor system that orchestrates the coordinated muscle contractions underlying movement. At the heart of this process lie the muscle spindles (MS), specialized receptors finely attuned to fluctuations in tension within intrafusal muscle fibres. The tension variation in the MS triggers a series of neuronal events including an initial depolarization of sensory type Ia afferents that subsequently causes the activation of motoneurons within the spinal cord1,2. This neuronal cascade culminates in the execution of muscle contraction, underscoring a presumed closed-loop mechanism between the musculoskeletal and nervous systems. By contrast, here we report the discovery of a new population of macrophages with exclusive molecular and functional signatures within the MS that express the machinery for synthesizing and releasing glutamate. Using mouse intersectional genetics with optogenetics and electrophysiology, we show that activation of MS macrophages (MSMP) drives proprioceptive sensory neuron firing on a millisecond timescale. MSMP activate spinal circuits, motor neurons and muscles by means of a glutamate-dependent mechanism that excites the MS. Furthermore, MSMP respond to neural and muscle activation by increasing the expression of glutaminase, enabling them to convert the uptaken glutamine released by myocytes during muscle contraction into glutamate. Selective silencing or depletion of MSMP in hindlimb muscles disrupted the modulation of the stretch reflex for force generation and sensory feedback correction, impairing locomotor strategies in mice. Our results have identified a new cellular component, the MSMP, that directly regulates neural activity and muscle contraction. The glutamate-mediated signalling of MSMP and their dynamic response to sensory cues introduce a new dimension to our understanding of sensation and motor action, potentially offering innovative therapeutic approaches in conditions that affect sensorimotor function.
Collapse
Affiliation(s)
- Yuyang Yan
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
- Faculty of Natural Sciences, Department of Life Sciences, Imperial College London, London, UK
| | - Nuria Antolin
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Luming Zhou
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Luyang Xu
- Faculty of Engineering, Department of Bioengineering, Imperial College London, London, UK
| | - Irene Lisa Vargas
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | | | - Guiping Kong
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Ilaria Palmisano
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Yi Yang
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Jessica Chadwick
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Franziska Müller
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Anthony M J Bull
- Faculty of Engineering, Department of Bioengineering, Imperial College London, London, UK
| | - Cristina Lo Celso
- Faculty of Natural Sciences, Department of Life Sciences, Imperial College London, London, UK
| | - Guido Primiano
- Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Serenella Servidei
- Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Carmelo Bellardita
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.
| | - Simone Di Giovanni
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK.
| |
Collapse
|
47
|
Zhang D, Wang X, Zhu L, Chen Y, Yang C, Zhong Z, Kong X, Nan J, Wang C, Hu H, Chen J, Shi P, Hu X, Zhu W, Wang J. TIMD4 hiMHCⅡ hi Macrophages Preserve Heart Function Through Retnla. JACC Basic Transl Sci 2025; 10:65-84. [PMID: 39906591 PMCID: PMC11788475 DOI: 10.1016/j.jacbts.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 02/06/2025]
Abstract
Genetic fate mapping confirmed the existence of the TIMD4hiMHCⅡhi macrophage subset and showed that they were resident macrophages with minimal input from peripheral monocytes. Further, single-cell RNA sequencing revealed that Retnla could serve as the signature gene for TIMD4hiMHCⅡhi macrophages. Administration of recombinant protein of the Retnla gene, RELMα, delayed the onset of heart failure, whereas either deletion of TIMD4hiMHCⅡhi macrophages or macrophage-specific loss of Retnla facilitated heart failure progression.
Collapse
Affiliation(s)
- Danyang Zhang
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xuanhao Wang
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Lianlian Zhu
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yuxing Chen
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chao Yang
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhiwei Zhong
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiangming Kong
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jinliang Nan
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chen Wang
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengxun Hu
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jinghai Chen
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng Shi
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Zhu
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jian’an Wang
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
48
|
Ferkel SAM, Holman EA, Sojwal RS, Rubin SJS, Rogalla S. Tumor-Infiltrating Immune Cells in Colorectal Cancer. Neoplasia 2025; 59:101091. [PMID: 39642846 DOI: 10.1016/j.neo.2024.101091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
Colorectal cancer encompasses a heterogeneous group of malignancies that differ in pathophysiological mechanisms, immune response and infiltration, therapeutic response, and clinical prognosis. Numerous studies have highlighted the clinical relevance of tumor-infiltrating immune cells among different types of colorectal tumors yet vary in cell type definitions and cell identification strategies. The distinction of immune signatures is particularly challenging when several immune subtypes are involved but crucial to identify novel intercellular mechanisms within the tumor microenvironment. In this review, we compile human and non-human studies on tumor-infiltrating immune cells and provide an overview of immune subtypes, their pathophysiological functions, and their prognostic role in colorectal cancer. We discuss how differentiating immune signatures can guide the development of immunotherapeutic targets and personalized treatment regimens. We analyzed comprehensive human protein biomarker profiles across the entire immune spectrum to improve interpretability and application of tumor studies and to ultimately enhance immunotherapy and advance precision medicine for colorectal cancer patients.
Collapse
Affiliation(s)
- Sonia A M Ferkel
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Elizabeth A Holman
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Raoul S Sojwal
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Samuel J S Rubin
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Stephan Rogalla
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA.
| |
Collapse
|
49
|
Wang YB, Li T, Wang FY, Yao X, Bai QX, Su HW, Liu J, Wang L, Tan RZ. The Dual Role of Cellular Senescence in Macrophages: Unveiling the Hidden Driver of Age-Related Inflammation in Kidney Disease. Int J Biol Sci 2025; 21:632-657. [PMID: 39781471 PMCID: PMC11705649 DOI: 10.7150/ijbs.104404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
Aging is a complex biological process that involves the gradual decline of cellular, tissue, and organ functions. In kidney, aging manifests as tubular atrophy, glomerulosclerosis, and progressive renal function decline. The critical role of senescence-associated macrophage in diseases, particularly kidney diseases, is increasingly recognized. During this process, macrophages exhibit a range of pro-damage response to senescent tissues and cells, while the aging of macrophages themselves also significantly influences disease progression, creating a bidirectional regulatory role between aging and macrophages. To explore this bidirectional mechanism, this review will elucidate the origin, characteristic, phenotype, and function of macrophages in response to the senescence-associated secretory phenotype (SASP), extracellular vesicles from senescent cells, and the senescence cell-engulfment suppression (SCES), particularly in the context of kidney disease. Additionally, it will discuss the characteristics of senescent macrophage, such as common markers, and changes in autophagy, metabolism, gene regulation, phagocytosis, antigen presentation, and exosome secretion, along with their physiological and pathological impacts on renal tissue cells. Furthermore, exploring therapies and drugs that modulate the function of senescent macrophages or eliminate senescent cells may help slow the progression of kidney aging and damage.
Collapse
Affiliation(s)
- Yi-bing Wang
- Department of Radiology, the Affiliated Hospital, Southwest Medical University, 646000 Luzhou, China
- Department of Medical Imaging, Southwest Medical University, 646000 Luzhou, China
| | - Tong Li
- Research Center of Integrated Traditional Chinese and Western Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, China
| | - Feng-yu Wang
- College of Integration of Traditional Chinese and Western Medicine, Southwest Medical University, 646000 Luzhou, China
| | - Xin Yao
- Department of Anesthesiology, Southwest Medical University, 646000 Luzhou, China
| | - Qiu-xiang Bai
- Research Center of Integrated Traditional Chinese and Western Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, China
| | - Hong-wei Su
- Department of Urology, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, China
| | - Jian Liu
- Department of Nephrology, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, China
| | - Li Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, China
| | - Rui-zhi Tan
- Research Center of Integrated Traditional Chinese and Western Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, China
| |
Collapse
|
50
|
Hermo L, Oliveira R, Dufresne J, Gregory M, Cyr DG. Basal and Immune Cells of the Epididymis: An Electron Microscopy View of Their Association. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:67-87. [PMID: 40301253 DOI: 10.1007/978-3-031-82990-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
The epididymis is a highly coiled duct divided into the initial segment, caput, corpus, and cauda regions. It is a pseudostratified epithelium consisting of principal, narrow, apical, basal, and clear cells. Circulating halo cells, identified as nonepithelial cells, monocytes/macrophages (M/M) and T-lymphocytes, in addition to dendritic cells and a resident population of M/M cells, also inhabit the epididymal epithelium. Using electron microscopy (EM), we characterized the ultrastructural features of each of these different cell types. Basal cells with stem cell characteristics suggest a role in sustaining the epithelium following injury and inflammation, as well as maintaining the steady state of the epithelium. Interestingly, a close morphological affiliation was noted between circulating M/M cells with basal cells and an intraepithelial resident M/M population of cells, as well as between T-lymphocytes and dendritic cells. The association of all these cell types with one another suggests complex interactions enabling the coordination of their functions related to maturation, protection, survival of sperm, and renewal of the epithelium.
Collapse
Affiliation(s)
- Louis Hermo
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Regiana Oliveira
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Julie Dufresne
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Mary Gregory
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Daniel G Cyr
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada.
| |
Collapse
|